EP2752191A1 - Compositions and methods using hdm2 antagonist and mek inhibitor - Google Patents

Compositions and methods using hdm2 antagonist and mek inhibitor Download PDF

Info

Publication number
EP2752191A1
EP2752191A1 EP13305008.8A EP13305008A EP2752191A1 EP 2752191 A1 EP2752191 A1 EP 2752191A1 EP 13305008 A EP13305008 A EP 13305008A EP 2752191 A1 EP2752191 A1 EP 2752191A1
Authority
EP
European Patent Office
Prior art keywords
compound
ray
additivity
synergy
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP13305008.8A
Other languages
German (de)
French (fr)
Inventor
Laurent Debussche
Jean-Paul Nicolas
Steve Rowley
James Watters
Fanny Windenberger
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi SA
Original Assignee
Sanofi SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanofi SA filed Critical Sanofi SA
Priority to EP13305008.8A priority Critical patent/EP2752191A1/en
Priority to PCT/US2014/010461 priority patent/WO2014107713A1/en
Publication of EP2752191A1 publication Critical patent/EP2752191A1/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • HDM2 negatively regulates p53 activity by binding to the transactivation domain of p53 and by acting as a p53-specific E3 ubiquitin ligase. In response to diverse stresses, p53 can induce cell cycle arrest and apoptosis.
  • Compound (1) is a selective antagonist of HDM2. ( WO2012/065022 ). Binding of Compound (1) to HDM2 disrupts the interaction between HDM2 and p53 thereby inhibiting the ability of HDM2 to bind to and inhibit p53.
  • Compound (4), N-((S)-2,3-dihydroxypropyl)-3-(2-fluoro-4-iodo-phenylamino)isonicotinamide (also referred to as Pimasertib, MSC1936369, or AS703026) is a novel, allosteric inhibitor of MEK. It possesses relatively high potency and selectivity, having no activity against 217 kinases or 90 non-kinase targets when tested at 10 ⁇ M. ( WO06/045514 ).
  • Tumor cells treated with inhibitors of MEK kinases typically respond via inhibition of phosphorylation of ERK, down-regulation of Cyclin D, induction of G1 arrest, and finally undergoing apoptosis.
  • the effect of MEK inhibition on tumor growth in selected mutant xenograft tumors that express mutant B-raf and selected Ras mutant tumors are described in D. B. Solit et al., Nature 2006; 439: 358-362 .
  • Predominant toxicities reported for MEK inhibitors in human patients include diarrhea, fatigue, rash, vomiting, nausea, and reversible visual disturbances.
  • Haura et al., Clin Cancer Res (2010) 16(8):2450-2457 Similar to epidermal growth factor receptor inhibitors, dermatologic toxicities such as papulopustular rash, xerosis, and pruritus are associated with MEK inhibitors.
  • dermatologic toxicities such as papulopustular rash, xerosis, and pruritus are associated with MEK inhibitors.
  • Baladula et al., Invest New Drugs (2011) 29:1114-1121 are associated with MEK inhibitors.
  • HDM2 antagonist and/or MEK inhibitor therapy that is made more efficacious without substantially increasing, or even maintaining or decreasing, the dosages of HDM2 antagonist and/or MEK inhibitor traditionally used in the art.
  • compositions for the treatment of patients having cancer and methods of using the compositions that employ an HDM2 antagonist in combination with an inhibitor of MEK pathway signaling.
  • methods for treating a subject having cancer comprising administering to the subject an effective amount of Compound (1): or a pharmaceutically acceptable salt thereof and Compound (4) or a pharmaceutically acceptable salt thereof.
  • Compound (1) and/or Compound (4) are administered with a pharmaceutically acceptable carrier.
  • Compound (1) and Compound (4) are administered in either separate or single dosage forms.
  • Compound (1) and Compound (4) are administered at different times.
  • Compound (1) and Compound (4) are administered at substantially the same time.
  • the effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  • a combination for use in treating a subject having cancer, the combination comprising a therapeutically effective amount of Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof.
  • the combination is in separate dosage forms, while in another embodiment, the combination is in a single dosage form.
  • a use of a combination comprising a therapeutically effective amount of Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in treatment of lymphoma is provided.
  • the combination is in separate dosage forms, while in another embodiment, the combination is in a single dosage form.
  • the therapeutically effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  • composition comprising Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof is provided.
  • the composition can further comprise a pharmaceutically acceptable carrier.
  • kits comprising: (A) Compound (1), or a pharmaceutically acceptable salt thereof; (B) Compound (4), or a pharmaceutically acceptable salt thereof; and (C) instructions for use.
  • the instructions are for the use of Compounds (1) and (4) in the treatment of cancer.
  • a therapeutically effective amount of Compound (1) and Compound (4) provided.
  • a therapeutically effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  • the cancer expresses mutant Nras.
  • the mutant Nras expressing cancer is melanoma.
  • the Nras mutant expressing cancer is bladder cancer.
  • the cancer expresses mutant Braf.
  • the Braf mutant expressing cancer is melanoma.
  • compositions and methods of use thereof for treatment of Nras mutant expressing bladder cancer or melanoma and for the treatment of mutant Braf expressing melanoma are provided herein that employ an HMD2 antagonist in combination with a MEK inhibitor.
  • the compositions provided herein afford better efficacy and/or potency than the single agent components of the compositions.
  • Compound (1) demonstrates synergistic activity in combination with Compound (4) in causing growth inhibition of Nras mutant expressing bladder cancer cells, mutant Nras expressing melanoma cells, and mutant Braf expressing melanoma cells.
  • Methods for treating a subject having cancer such as Nras mutant expressing bladder cancer or melanoma and for the treatment of mutant Braf expressing melanoma are provided.
  • the methods comprise administering to the patient a therapeutically effective amount of an HDM2 antagonist and MEK inhibitor as further described herein.
  • the methods and compositions comprise a HDM2 antagonist having the formula (1):
  • Compound (1) The compound according to formula (1) is referred to herein as "Compound (1)".
  • the preparation and properties of Compound (1) are provided in, for example, International Patent Publication No. WO2012/065022 , particularly Example 11, p. 169-171 therein. The entire contents of WO2012/065022 are incorporated herein by reference.
  • the methods and compositions comprise a MEK inhibitor having the formula (4):
  • Compound (4) The compound according to formula (4) is referred to herein as "Compound (4)".
  • the preparation and properties of Compound (4), also referred to as Pimasertib, are provided in, for example, International Patent Publication No. WO06/045514 , particularly Example 115 and Table 1 therein. The entire contents of WO06/045514 are incorporated herein by reference.
  • the compounds described herein are unsolvated.
  • one or both of the compounds used are in solvated form.
  • the solvate can be any ofpharmaceutically acceptable solvent, such as water, ethanol, and the like.
  • the presence of a solvate or lack thereof does not have a substantial effect on the efficacy of Compounds (1) or (4) described above.
  • the Compounds (1) and (4) are depicted in their neutral forms, in some embodiments, these compounds are used in a pharmaceutically acceptable salt form.
  • the salt can be obtained by any of the methods well known in the art.
  • a "pharmaceutically acceptable salt" of the compound refers to a salt that is pharmaceutically acceptable and that retains pharmacological activity.
  • Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, as well as those salts formed with organic acids, such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-
  • Compound (1) is administered simultaneously with Compound (4).
  • Simultaneous administration typically means that both compounds enter the patient at precisely the same time.
  • simultaneous administration also includes the possibility that Compound (1) and Compound (4) enter the patient at different times, but the difference in time is sufficiently miniscule that the first administered compound is not provided the time to take effect on the patient before entry of the second administered compound.
  • Such delayed times typically correspond to less than 1 minute, and more typically, less than 30 seconds.
  • simultaneous administration can be achieved by administering a solution containing the combination of compounds.
  • simultaneous administration of separate solutions one of which contains the Compound (1) and the other of which contains Compound (4), can be employed.
  • simultaneous administration can be achieved by administering a composition containing the combination of compounds.
  • Compound (1) and Compound (4) are not simultaneously administered.
  • the Compound (1) is administered before Compound (4).
  • Compound (4) is administered before Compound (1).
  • the time difference in non-simultaneous administrations can be greater than 1 minute, and can be, for example, precisely, at least, up to, or less than 5 minutes, 10 minutes, 15 minutes, 30 minutes, 45 minutes, 60 minutes, two hours, three hours, six hours, nine hours, 12 hours, 24 hours, 36 hours, or 48 hours.
  • the first administered compound is provided time to take effect on the patient before the second administered compound is administered. Generally, the difference in time does not extend beyond the time for the first administered compound to complete its effect in the patient, or beyond the time the first administered compound is completely or substantially eliminated or deactivated in the patient.
  • one or both of the Compounds (1) and (4) are administered in a therapeutically effective (i.e., therapeutic) amount or dosage.
  • a “therapeutically effective amount” is an amount of Compound (1) or Compound (4) that, when administered to a patient by itself, effectively treats the cancer (for example, inhibits tumor growth, stops tumor growth, or causes tumor regression).
  • An amount that proves "therapeutically effective amount" in a given instance, for a particular subject, may not be effective for 100% of subjects similarly treated for the disease or condition under consideration, even though such dosage is deemed a "therapeutically effective amount” by skilled practitioners.
  • the amount of the compound that corresponds to a therapeutically effective amount is affected by the type of cancer, stage of the cancer, the age of the patient being treated, and other facts.
  • one or both of the Compounds (1) and (4) are administered in a sub-therapeutically effective amount or dosage.
  • a sub-therapeutically effective amount is an amount of Compound (1) or (4) that, when administered to a patient by itself, does not completely inhibit over time the biological activity of the intended target.
  • the combination of Compounds (1) and (4) should be effective in treating, for example mutant Nras expressing melanoma mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  • a sub-therapeutic amount of Compound (4) can be an effective amount if, when combined with Compound (1), the combination is effective in the treatment of mutant Nras expressing melanoma mutant Nras expressing bladder cancer, or mutant Braf expressing melanoma.
  • synergistic effect refers to action of two agents such as, for example, Compound (1) and Compound (4), producing an effect, for example, slowing the symptomatic progression of cancer or symptoms thereof, which is greater than the simple addition of the effects of each drug administered by themselves.
  • a synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emax equation ( Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet. 6: 429-453 (1981 )), the equation of Loewe additivity ( Loewe, S.
  • the combination of compounds can inhibit cancer growth.
  • the combination achieves cancer stasis. In some embodiments, the combination achieves substantial or complete cancer regression.
  • the term "about” generally indicates a possible variation of no more than 10%, 5%, or 1% of a value. For example, “about 25 mg/kg” will generally indicate, in its broadest sense, a value of 22.5-27.5 mg/kg, i.e., 25 ⁇ 10 mg/kg.
  • the amounts of Compounds (1) and (4) should result in the effective treatment of a cancer
  • the amounts, when combined, are preferably not excessively toxic to the patient (i.e., the amounts are preferably within toxicity limits as established by medical guidelines).
  • a limitation on the total administered dosage is provided.
  • the amounts considered herein are per day; however, half-day and two-day or three-day cycles also are considered herein.
  • a daily dosage such as any of the exemplary dosages described above, is administered once, twice, three times, or four times a day for three, four, five, six, seven, eight, nine, or ten days.
  • a shorter treatment time e.g., up to five days
  • a longer treatment time e.g., ten or more days, or weeks, or a month, or longer
  • a once- or twice-daily dosage is administered every other day.
  • each dosage contains both Compound (1) and Compound (4) to be delivered as a single dosage, while in other embodiments, each dosage contains either Compound (1) or Compound (4) to be delivered as separate dosages.
  • Examples of types of cancers to be treated with the present invention include, for example, mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  • the subject considered herein is typically a human, for example a human patient.
  • the subject can be any mammal for which cancer treatment is desired.
  • the methods described herein can be applied to both human and veterinary applications.
  • treating indicates that the method has, at the least, mitigated abnormal cellular proliferation.
  • the method can reduce the rate of cancer growth in a patient, or prevent the continued growth of a cancer, or even reduce the overall reach of the cancer
  • Compounds (1) and (4), or their pharmaceutically acceptable salts or solvate forms, in pure form or in an appropriate pharmaceutical composition, can be administered via any of the accepted modes of administration or agents known in the art.
  • the compounds can be administered, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally.
  • the dosage form can be, for example, a solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, pills, soft elastic or hard gelatin capsules, powders, solutions, suspensions, suppositories, aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
  • a particular route of administration is oral, particularly one in which a convenient daily dosage regimen can be adjusted according to the degree of severity of the disease to be treated.
  • a composition that comprises Compound (1) and Compound (4).
  • the composition is in the form of a solid (e.g., a powder or tablet) including Compound (1) and Compound (4) in solid form, and optionally, one or more auxiliary (e.g., adjuvant) or pharmaceutically active compounds in solid form.
  • the composition further includes any one or combination of pharmaceutically acceptable carriers (i.e., vehicles or excipients) known in the art, thereby providing a liquid dosage form.
  • Compound (1) and Compound (4) of the combination therapy can be administered in a single unit does or separate dosage forms. Accordingly, the pharmaceutically acceptable carriers and excipients described throughout the application can be combined with Compounds (1) and (4) in a single unit dose, as well as individually combined with Compounds (1) and (4) when these compounds are administered separately.
  • Auxiliary and adjuvant agents may include, for example, preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents.
  • Prevention of the action of microorganisms is generally provided by various antibacterial and antifungal agents, such as, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Isotonic agents such as sugars, sodium chloride, and the like, may also be included.
  • Prolonged absorption of an injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • the auxiliary agents also can include wetting agents, emulsifying agents, pH buffering agents, and antioxidants, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylated hydroxytoluene, and the like.
  • Dosage forms suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid
  • binders as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia
  • humectants as for example, glycerol
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate
  • solution retarders as for example paraffin
  • absorption accelerators as for example, quaternary
  • Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well-known in the art. They can contain pacifying agents and can be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds also can be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., the HDM2 antagonist or the MEK inhibitor compound described herein, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3- butyleneglycol, dimethyl formamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfury
  • Suspensions in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • suspending agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds described herein with, for example, suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • Dosage forms for topical administration may include, for example, ointments, powders, sprays, and inhalants.
  • the active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as can be required.
  • Ophthalmic formulations, eye ointments, powders, and solutions also can be employed.
  • the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of the compounds described herein, or a pharmaceutically acceptable salt thereof, and 99% to 1% by weight of a pharmaceutically acceptable excipient.
  • the composition will be between about 5% and about 75% by weight of a compounds described herein, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
  • the composition includes one or more other anti-cancer compounds.
  • administered compositions can comprise standard of care agents for the type of cancers selected for treatment.
  • kits are provided. Such kits can comprise package(s) comprising compounds or compositions as described herein. In one embodiment, kits comprise Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4) or a pharmaceutically acceptable salt thereof.
  • packaging means any vessel containing compounds or compositions presented herein.
  • the package can be a box or wrapping.
  • Packaging materials for use in packaging pharmaceutical products are well-known to those of skill in the art. Examples of pharmaceutical packaging materials include, but are not limited to, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • the kit also can contain items that are not contained within the package but are attached to the outside of the package, for example, pipettes.
  • Kits can contain instructions for administering compounds or compositions of the invention to a patient. Kits also can comprise instructions for approved uses of compounds herein by regulatory agencies, such as the United States Food and Drug Administration. Kits also can contain labeling or product inserts for the inventive compounds. The package(s) and/or any product insert(s) may themselves be approved by regulatory agencies.
  • the kits can include compounds in the solid phase or in a liquid phase (such as buffers provided) in a package.
  • the kits also can include buffers for preparing solutions for conducting the methods, and pipettes for transferring liquids from one container to another.
  • Compound (5) also referred to as Trametinib, GSK1120212, GSK212, and JTP-74057
  • Trametinib also referred to as Trametinib, GSK1120212, GSK212, and JTP-74057
  • Compound (7) also referred as PD0325901 (N-(2,3-dihydroxy-propoxy)-3,4-difluoro-2-(2-fluoro-4-iodo-phenylamino)-benzamide) are provided in, for example, in published PCT application WO2002006213 , particularly Example 85 therein, the contents of which are incorporated herein by reference.
  • Compound (8) also referred as also referred to as 4-[2-(2-chloro-4-fluorophenylamino)-5-methylpyrimidin-4-yl]-N-[1(S)-(3-chlorophenyl)-2-hydroxyethyl]-1H-pyrrole-2-carboxamide are provided in, for example, in Aronov, et al, J.Med. Chem. (2009) 52:6362-6368 , the contents of which are incorporated herein by reference.
  • RO-5126766 N-[3-fluoro-4-[4-methyl-2-oxo-7-(pyrimidin-2-yloxy)-2H-1-benzopyran-3-ylmethyl]pyridin-2-yl]-N'-methylsulfamide
  • RO-5126766 N-[3-fluoro-4-[4-methyl-2-oxo-7-(pyrimidin-2-yloxy)-2H-1-benzopyran-3-ylmethyl]pyridin-2-yl]-N'-methylsulfamide
  • Compound (10) also referred to as (6-methoxy-7-[3-(4-morpholinyl)propoxy]-4-(4-phenoxyphenylamino)quinoline-3-carbonitrile), are provided in, for example, in Zhang, et al, Bioor. Med. Chem. Let. (2000) 10:2825-2828 , the contents of which are incorporated herein by reference.
  • the combinations of HDM2-p53 inhibitor and MEK inhibitor were examined in a panel of twenty cell lines (Table 2).
  • the panel of cell lines included cell lines that were induced to undergo apoptosis upon exposure to Compound (1) (apoptosis), as well as cell lines where exposure to Compound (1) resulted in inhibition of cell proliferation but not apoptosis (stasis), and cell lines that did not undergo apoptosis or experience stasis upon exposure to Compound (1) (resistant).
  • ATP levels were used as a measure of cell number (cell growth). At the time of treatment, ATP levels were measured in an untreated set of assay plates (Tzero plates). ATP levels were measured by adding ATPLite (Perkin Elmer) according to manufacturer's instructions. The Tzero plates were read using ultra-sensitive luminescence on Envision Plate Readers. Assay plates receiving treatment were incubated with compound for 72 h at 37° C with 5% CO 2 . After 72 h, plates were developed for endpoint analysis using ATPLite.
  • GI Growth Inhibition
  • GI was calculated by applying the following test and equation: If T ⁇ V 0 : 100 * 1 - T - V 0 / V 0 If T ⁇ V 0 : 100 * 1 - T - V 0 / V - V 0
  • T is the signal measure for a test article
  • V is the vehicle-treated control measure
  • V 0 is the vehicle control measure at time zero.
  • This formula is derived from the Growth Inhibition calculation used in the National Cancer Institute's NCI-60 high throughput screen, as described on the world wide web at dtp.nci.nih.gov/branches/btb/ivclsp.html.
  • a GI reading of 0% represents no growth inhibition-that is, cells treated with compound had similar levels of ATP as cells treated with vehicle and measured at 72 hours.
  • a GI of 100% represents complete growth inhibition-that is, cells treated with compound had similar levels of ATP as cells treated with vehicle and measured at T0.
  • a GI of 100% where cell numbers have not increased during the treatment period is indicative of a cytostatic effect for compounds reaching a plateau at this effect level.
  • a GI of 200% represents complete death of all cells in the culture well which is indicative of an apoptotic effect.
  • the dose-response curve was measured with each HDM2-p53 antagonist and each MEK inhibitor as single agents and in pair wise combinations. Using the potency values from the single agent dose-response curves, the compounds were assigned to different groups of similar potency. Multiple doses above and below the EC 50 of a given HDM2-p53 antagonist or MEK inhibitor were measured.
  • ATP levels were measured by fluorescence assay as described above. ATP levels are proportional to the number of remaining live cells.
  • This raw data was then normalized in two steps. First, background subtraction was performed by subtracting a mean intensity of negative control wells in which cells were dosed with dimethyl sulfoxide (DMSO). Second, a dynamic range normalization was performed by dividing the background-subtracted data by the mean intensity of positive control wells in which cells were dosed with Staurosporine. The resulting normalized intensity, now with plate-specific effects removed, was used to calculate the Growth Inhibition measure described above.
  • DMSO dimethyl sulfoxide
  • a dose response matrix was constructed where one axis corresponded to one of the three HDM2-p53 antagonists, and the other axis corresponded to one of the MEK inhibitors.
  • the high doses were approximately the given compound's EC 90 and were diluted by a constant factor to generate the lower dosage levels.
  • the lowest dose was 0, i.e., the edges of the matrix correspond to another measure of single-agent behavior of each of the compounds.
  • compounds were combined with themselves in a self-cross analysis for each cell line as described below.
  • Synergy in general means that two or more compounds, in a given biological context, have a combined effect that is significantly greater than would be expected from the combinations of their single-agent effects.
  • Synergy can be measured in a variety of ways; the methods for measuring synergy involve stating a null hypothesis about the expected single-agent effects, and computing a summary score of the differences between observations and the null hypothesis. A variety of synergy scores in these three broad families were considered.
  • Loewe Synergy where the null hypothesis is that the two compounds being evaluated are actually the same compound (a self-cross).
  • the effect of combining the two compounds is a function of the sum of concentrations of the two compounds.
  • Significant synergy means the compounds have different effects which work together to create an effect.
  • Gaddum Synergy also known as Highest Single Agent Synergy, where the null hypothesis is that there is competitive binding between the two compounds for the same target, and only the compound with the highest binder affinity causes an effect.
  • Significant synergy means that something other than competitive binding is occurring, in which the compounds produce their combined effect in independent ways.
  • FIG. 1 shows a table of the mean synergy score for Compounds (1) and (2) in combination with the indicated MEK inhibitor in the indicated cell lines.
  • the grey color shows the maximum FDR with Compound (1) and Compound (2) in the indicated cell line.
  • MEK inhibitors (6), (7), (8), (9), and (10) showed statistically significant Loewe synergy with both Compound (1) and Compound (2) at an FDR ⁇ 5% in some cell lines.
  • FIG. 2 shows effect of the combination of Compound (1) and Compound (6) on C32 cells.
  • FIG. 2A shows the mean synergy scores of the combination of Compounds (1) and (6) at the indicated concentrations.
  • FIG. 2B shows percent growth inhibition as a function of concentration of Compound (1), where Compound (6) is not present or is present at 1.1 uM.
  • Compound (1) and Compound (6) induce cytostasis of C32 cells when used as single agents.
  • the combination of Compound (1) and (6) induced significant cell death.
  • FIG. 3 shows effect of the combination of Compound (1) and Compound (6) on MV4-11 cells.
  • FIG. 2A shows the mean synergy scores of the combination of Compounds (1) and (6) at the indicated concentrations.
  • FIG. 2B shows percent growth inhibition as a function of concentration of Compound (1), where Compound (6) is not present or is present at 3.3 uM.
  • Compound (1) induces cytotoxicity in MV4-11 cells and Compound (6) induces cytostasis of MV4-11 cells when used as single agents.
  • the combination of Compound (1) and (6) resulted in a cytotoxicity at lower concentrations of Compound (1).
  • FIG. 4 shows effect of the combination of Compound (1) and Compound (6) on A375 cells.
  • FIG. 4A shows the mean synergy scores of the combination of Compounds (1) and (6) at the indicated concentrations.
  • FIG. 4B shows an isobologram of growth inhibition of A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (6), compared to additive growth inhibition.
  • the combination of Compounds (1) and (6) displayed synergy in inhibition of A375 cell proliferation.
  • the Loewe Synergy Score for the treatment of A375 cells with the combination of Compounds (1) and (5) is 2.7.
  • FIG. 5 shows effect of the combination of Compound (1) and Compound (5) on Vemurafenib resistant A375 cells.
  • FIG. 5A shows the mean synergy scores of the combination of Compounds (1) and (5) at the indicated concentrations.
  • FIG. 5B shows an isobologram of growth inhibition of Vemurafenib resistant A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (5), compared to additive growth inhibition.
  • the combination of Compounds (1) and (5) displayed synergy in inhibition of Vemurafenib resistant A375 cell proliferation.
  • the Loewe Synergy Score for the treatment of Vemurafenib resistant A375 cells with the combination of Compounds (1) and (5) is 2.7.
  • Cells were seeded in 384 well microplates in 47.5 ⁇ L of culture medium as described in Table 4 and incubated for 4 hours at 37° C with 5 % CO2. After incubation, 2.5 ⁇ L of a 20x concentrated solution of individual test compounds (for single agent tests) or mixtures of test compounds (for combination tests) were added (see "Cell Treatment” section). The microplates are returned to the incubator for 96 h.
  • a mix of lysis reagent and luciferase/luciferin was added (CellTiterGlo, Promega) in each well, the microplates were incubated for 1 h at room temperature and the luminescence emitted was measured with a Microbeta Trilux counter (PerkinElmer).
  • a combination dose response matrix was generated with Compound (1) and Compound (4) or Compound (5) in a 384 well plate.
  • Compound (1) was diluted to generate 14 concentrations and Compound (4) (or Compound (5)) was diluted to generate 18 concentrations. All combinations of concentrations were made in a 14x18 matrix (252 wells). Two identical plates were prepared, allowing duplicate determination of the effect of each combination.
  • ray design was used to validate findings of synergy. Compound (1) and Compound (4) or Compound (5) were evaluated at several fixed proportions of the compounds in combination. ( R. Straetemans, Biometrical journal 47 -2005 ). The ray design included one ray for each single agent and 19 combination rays. The ray with Compound (1) alone had 14 concentrations of Compound (1), the ray with Compound (4) or Compound (5) alone had 18 concentrations of Compound (4) or Compound (5), and the combination rays had between 7 and 14 concentrations of each compound.
  • Absolute ICX is defined as the concentration of compound where 1% is equal to X%. This measurement allows determining the potential synergistic combinations using the statistical method described hereunder.
  • Y ijk is the percentage of inhibition for the k th replicate of the j th concentration in the i th ray
  • Conc ij is the j th mixture concentration (sum of the concentrations of compound A and compound B) in the i th ray
  • i is the minimum effect obtained from i th ray
  • Emax i is the maximum effect obtained from i th ray
  • IC50 i is the IC50 obtained from i th ray
  • m i is the slope of the curve adjusted with data from i th ray
  • ⁇ ijk is the residual for the k th replicate of the j th concentration in the i th ray, ⁇ ijk ⁇ N(0, ⁇ 2 )
  • Emin, Emax and/or slope were shared whenever it was possible without degrading the quality of the fit.
  • IC50 or IC40 for compounds with a low activity
  • Additivity for a given Ray was defined as a 95% confidence interval of the Ki that included 1.
  • Significant synergy for a given Ray was defined as a 95% confidence interval of the Ki having an upper bound of less than 1.
  • Significant antagonism was defined as a 95% confidence interval of the Ki having a lower bound of higher than 1.
  • the isobolograms shown in FIGs. 6-17 allow visualization of the position of each Ray.
  • the line shown in FIGs. 6-17 connects the point (0,1) to the point (1,0). All rays below the line correspond to potential synergy, whereas rays above the line correspond to potential antagonism.
  • the x-axis is [Compound (1)]/IC50[Compound (1)] and the y-axis is [Compound (4)]/IC50[Compound(4)] (FIG. A) or [Compound (5)]/IC50[Compound(5)] (FIG. B).
  • the x-axis is [Compound (1)]/IC40[Compound (1)] and the y-axis is [Compound (4)]/IC40[Compound(4)] ( FIG. 10A ) or [Compound (5)]/IC40[Compound(5)] ( FIG. 10B ).
  • Compound (1) shows a trend toward antagonism with Compound (4) and additivity with Compound (5) in an Nras mutant melanoma cell line (C8161).
  • Table 5 shows the IC50 estimations for each compound and the relative potency of Compounds (1) and (4) on C8161 cells when used as single agents.
  • Table 5 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) N1 (IC50) 7.74E+01 [6.16E+01 ; 9.72E+01] 1.89E+02 [1.38E+02 ; 2.60E+02] 0.41 N2 (IC50) N2 (IC50) 1.63E+02 [9.31E+01;2.84E+02] 2.87E+02 [2.24E+02 ; 3.68E+02] 0.57
  • Table 6 shows the interaction characterization of Compounds (1) and (4) in C8161 cells.
  • Table 6 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 5 0.05 1.2284 [0.8518 ; 1.7716] Additivity N1 Ray 5 0.07 1.3302 [0.7936 ; 2.2295] Additivity N2 Ray 6 0.15 1.4596 [0.9350 ; 2.2784] Additivity N1 Ray 6 0.20 1.4464 [0.8138 ; 2.5707] Additivity N2 Ray 7 0.35 1.4928 [1.0483 ; 2.1259] Antagonism N1 Ray 7 0.42 1.3625 [0.9226 ; 2.0121] Additivity N2 Ray 8 0.64 1.1827 [0.6624 ; 2.1119] Additivity N1 Ray 8 0.71 1.2192 [0.8524 ; 1.7439] Additivity N2 Ray 9 0.85 1.2099 [0.3842 ; 3.8105] Additivity N1 Ray 9 0.89 1.2529 [0.8711 ;
  • Table 7 shows the IC50 estimations for each compound and the relative potency of Compounds (1) and (5) on C8161 cells when used as single agents.
  • Table 7 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 1.42E+02 [1.15E+02 ; 1.75E+02] 2.80E+01 [2.13E+01 ; 3.68E+01] 5.07 N2 (IC50) 9.19E+01 [6.52E+01 ; 1.29E+02] 3.00E+01 [1.83E+01 ; 4.92E+01] 3.06
  • Table 8 shows the interaction characterization of Compounds (1) and (4) in C8161 cells.
  • Table 8 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 7 0.06 0.7853 [0.4884 ; 1.2627] Additivity N3 Ray 6 0.09 1.0619 [0.4659 ; 2.4204] Additivity N1 Ray 8 0.16 0.7728 [0.4591 ; 1.3008] Additivity N3 Ray 7 0.25 1.3380 [0.6550 ; 2.7333] Additivity N1 Ray 9 0.40 1.0063 [0.6789 ; 1.4917] Additivity N3 Ray 8 0.52 1.0316 [0.5872 ; 1.8122] Additivity N1 Ray 10 0.66 0.9705 [0.6897 ; 1.3655] Additivity N3 Ray 9 0.77 1.0863 [0.6502 ; 1.8150] Additivity N1 Ray 11 0.87 0.7584 [0.5434 ; 1.0584] Additivity N3 Ray 10 0.92 0.7807 [0.4402
  • FIG. 6 shows an isobologram of growth inhibition of C8161 cells treated with Compound (1) in combination with Compound (4) ( FIG. 6A ) or with Compound (5) ( FIG. 6B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) showed a trend toward antagonsism and Compound (1) in combination with (5) showed an additive effect on C8161 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in HCT116 Cells
  • Table 9 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on HCT116 cells.
  • Table 9 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 2.96E+02 [1.41E+02 ; 3.63E+02] 1.50E+02 [1.20E+02 ; 1.86E+02] 1.98 N2 (IC50) 2.96E+02 [2.33E+02 ; 3.77E+02] 1.59E+02 [1.21E+02 ; 2.09E+02] 1.86 N3 (IC50) 5.30E+02 [3.56E+02 ; 7.89E+02] 4.21E+01 [2.91E+01 ; 6.10E+02] 12.58 N4 (IC50) 4.92E+02 [3.07E+02 ; 7.87E+02] 6.76E+01 [4.18E+01 ; 1.10E+02] 7.28
  • Table 10 shows the interaction characterization of Compounds (1) and (4) in HCT116 cells.
  • Table 10 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N4 Ray 7 0.04 1.0245 [0.4100 ; 2.5596] Additivity N1 Ray 6 0.05 0.5864 [0.3866 ; 0.8896] Synergy N2 Ray 6 0.05 0.7371 [0.3785 ; 1.4354] Additivity N3 Ray 8 0.07 1.6414 [0.8931 ; 3.0168] Additivity N4 Ray 8 0.12 0.9923 [0.4493 ; 2.1919] Additivity N1 Ray 7 0.13 0.5293 [0.3563 ; 0.7864] Synergy N2 Ray 7 0.14 1.7545 [1.1893 ; 2.5883] Antagonism N3 Ray 9 0.21 1.0670 [0.6146 ; 1.8524] Additivity N4 Ray 9 0.31 0.6700 [0.3251 ; 1.3808] Additivity N1 Ray 8 0.34 0.5892 [0.4156
  • Table 11 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on HCT116 cells.
  • Table 11 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) N1 (IC50) 3.66E+02 [2.95E+02 ; 4.56E+02] 1.37E+01 [1.08E+01 ; 1.73E+01] 26.84 N2 (IC50) 2.43E+02 [2.01E+03 ; 2.95E+02] 1.04E+01 [8.38E+00 ; 1.28E+01] 23.49 N3 (IC50) 6.42E+02 [4.90E+02 ; 8.42E+02] 1.96E+01 [1.49E+01 ; 2.59E+01] 32.75
  • Table 12 shows the interaction characterization of Compounds (1) and (5) in HCT116 cells.
  • Table 12 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N3 Ray 8 0.03 0.4099 [0.2339 ; 0.7186] Synergy N1 Ray 8 0.04 0.7768 [0.5208 ; 1.1586] Additivity N2 Ray 8 0.04 0.7355 [0.5100 ; 1.0608] Additivity N3 Ray 9 0.09 0.4684 [0.2814 ; 0.7798] Synergy N1 Ray 9 0.11 0.8389 [0.5625 ; 1.2512] Additivity N2 Ray 9 0.12 0.8544 [0.6081 ; 1.2005] Additivity N3 Ray 10 0.23 0.4153 [0.2621 ; 0.6580] Synergy N1 Ray 10 0.27 0.6508 [0.4369 ; 0.9696] Synergy N2 Ray 10 0.30 0.6879 [0.4947 ; 0.9566] Synergy N3 Ray 11 0.50 0.5036 [0.3
  • FIG. 7 shows an isobologram of growth inhibition of HCT116 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 7A ) or Compound (5) ( FIG. 7B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) or (5) showed a synergistic effect on HCT116 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in an Nras mutant melanoma cell line, SK-MEL-103.
  • Table 13 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on SK-MEL-103 cells.
  • Table 13 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 2.60E+02 [2.11E+02 ; 3.20E+02] 2.85E+01 [2.30E+01 ; 3.53E+01] 9.1 N2 (IC50) 3.41E+02 [2.62E+02 ; 4.43E+02] 3.16E+01 [2.38E+01 ; 4.18E+01] 10.8 N3 (IC50) 2.44E+02 [2.10E+02 ; 2.83E+02] 3.40E+01 [2.77E+01 ; 4.17E+01] 7.2
  • Table 14 shows the interaction characterization of Compounds (1) and (4) in SK-MEL-103 cells.
  • Table 14 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 7 0.03 0.5733 [0.3874 ; 0.8483] Synergy N3 Ray 7 0.04 0.7592 [0.5434 ; 1.0606] Additivity N2 Ray 8 0.08 0.9332 [0.5787 ; 1.5047] Additivity N1 Ray 8 0.10 0.7038 [0.4942 ; 1.0022] Additivity N3 Ray 8 0.12 0.8963 [0.6406 ; 1.2542] Additivity N2 Ray 9 0.24 0.7623 [0.4920 ; 1.1811] Additivity N1 Ray 9 0.27 0.6873 [0.4967 ; 0.9511] Synergy N3 Ray 9 0.32 0.5607 [0.4173 ; 0.7534] Synergy N2 Ray 10 0.48 0.6409 [0.4261 ; 0.9640] Synergy N1 Ray 10 0.52
  • Table 15 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on SK-MEL-103 cells.
  • Table 15 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 3.60E+02 [2.82E+02 ; 4.60E+02] 4.83E+00 [3.79E+00 ; 6.16E+00] 74.6 N2 (IC50) 3.63E+02 [3.00E+02 ; 4.39E+02] 4.58E+00 [3.79E+00 ; 5.54E+00] 79.1 N3 (IC50) 3.59E+02 [2.82E+02 ; 4.58E+02] 4.47E+00 [3.50E+00 ; 5.73E+00] 80.3
  • Table 16 shows the interaction characterization of Compounds (1) and (5) in SK-MEL-103 cells.
  • Table 16 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N3 Ray 9 0.04 0.7550 [0.4853 ; 1.1747] Additivity N2 Ray 9 0.04 0.6809 [0.4798 ; 0.9664] Synergy N1 Ray 9 0.04 0.9716 [0.5780 ; 1.6332] Additivity N3 Ray 10 0.11 0.7894 [0.527 ; 1.1816] Additivity N2 Ray 10 0.11 0.6393 [0.4555 ; 0.8974] Synergy N1 Ray 10 0.12 0.5086 [0.3376 ; 0.7662] Synergy N3 Ray 11 0.29 0.5741 [0.4014 ; 0.8209] Synergy N2 Ray 11 0.30 0.5522 [0.4075 ; 0.7485] Synergy N1 Ray 11 0.31 0.5301 [0.3522 ; 0.7979] Synergy N3 Ray 12 0.
  • FIG. 8 shows an isobologram of growth inhibition of SK-MEL-103 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 8A ) or with Compound (5) ( FIG. 8B ), compared to additive growth inhibition.
  • the Compound (1) in combination with Compound (4) or Compound (5) showed a synergistic effect on SK-MEL-103 cells.
  • Compound (1) shows a trend to synergy with Compounds (4) and (5) in UACC-62 cells.
  • Table 17 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on UACC-62 cells.
  • Table 17 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 2.05E+02 [1.51E+02 ; 2.78E+02] 9.87E+00 [7.75E+00; 1.26E+01] 20.8 N2 (IC50) 3.44E+02 [2.66E+02 ; 4.45E+02] 5.16E+00 [4.17E+00 ; 6.39E+00] 66.72 N3 (IC50) 1.07E+02 [4.05E+01;2.84E+02] 9.29E+00 [6.57E+00 ; 1.31E+01] 11.55
  • Table 18 shows the interaction characterization of Compounds (1) and (4) in UACC-62 cells.
  • Table 18 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 8 0.05 0.6078 [0.3961 ; 0.9326] synergy N2 Ray 9 0.05 1.2738 [0.8904 ; 1.8222] additivity N3 Ray 8 0.08 1.0897 [0.5725 ; 2.0740] additivity N2 Ray 10 0.13 1.0951 [0.7723 ; 1.5529] additivity N1 Ray 9 0.14 0.5185 [0.3286 ; 0.8182] synergy N3 Ray 9 0.22 0.8687 [0.4733 ; 1.5945] additivity N1 Ray 10 0.32 0.6805 [0.4350 ; 1.0647] additivity N2 Ray 11 0.33 0.7797 [0.5547 ; 1.0960] additivity N3 Ray 10 0.46 0.4797 [0.2268 ; 1.0147] additivity N2 Ray 12 0.60 0.6365 [0.4
  • Table 19 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on UACC-62 cells.
  • Table 19 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 1.95E+02 [1.54E+02 ; 2.47E+02] 8.35E-01 [6.76E-01 ; 1.03E+00] 233.36 N2 (IC50) 1. 12E+02 [7.23E+01 ; 1.72E+02] 1.14E+00 [8.02E-01 ; 1.63E+00] 98.25 N3 (IC50) 1.32E+02 [9.87E+01 ; 1.77E+02] 1.03E+00 [8.06E-01 ; 1.31E+00] 128.15
  • Table 20 shows the interaction characterization of Compounds (1) and (5) in UACC-62 cells.
  • Table 20 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 9 0.03 0.4761 [0.2605 ; 0.8702] Synergy N1 Ray 10 0.04 0.8046 [0.5605 ; 1.1551] Additivity N3 Ray 10 0.07 0.4425 [0.2921 ; 0.6704] synergy N2 Ray 10 0.09 0.4787 [0.2705 ; 0.8470] Synergy N1 Ray 11 0.12 0.6280 [0.4411 ; 0.8942] Synergy N3 Ray 11 0.21 0.6226 [0.4178 ; 0.9278] synergy N2 Ray 11 0.25 0.4913 [0.2781 ; 0.8680] Synergy N1 Ray 12 0.30 0.8162 [0.5788 ; 1.1510] Additivity N3 Ray 12 0.44 0.7460 [0.4982 ; 1.1173] additivity N2 Ray 12 0.51 0.
  • FIG. 9 shows an isobologram of growth inhibition of UACC-62 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 9A ) or Compound (5) ( FIG. 9B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) or Compound (5) showed a trend toward synergy on UACC-62 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in HT-1197 cells.
  • Table 21 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on HT-1197 cells.
  • Table 21 Experiment Compound 1 Compound 4 Relative potency N1 (IC40) 3.68E+02 [2.04E+02 ; 6.63E+02] 3.81E+02 [1.98E+02 ; 7.35E+02] 0.97 N2 (IC40) 4.10E+02 [2.10E+02 ; 8.00E+02] 7.30E+01 [3.79E+01 ; 1.41E+02] 5.61 N3 (IC40) 6.07E+02 [1.93E+02 ; 1.91E+03] 1.30E+02 [5.25E+01 ; 3.20E+02] 4.68
  • Table 22 shows interaction characteristics of Compounds (1) and (4) in HT-1197 cells.
  • Table 22 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 7 0.05 0.3150 [0.1005 ; 0.9873] Synergy N3 Ray 7 0.06 0.7615 [0.1413 ; 4.1039] Additivity N1 Ray 6 0.09 0.0724 [0.0242 ; 0.2164] Synergy N2 Ray 8 0.15 0.4372 [0.1668 ; 1.1462] Additivity N3 Ray 8 0.18 0.4460 [0.0992 ; 2.0060] Additivity N1 Ray 7 0.24 0.0285 [0.0110 ; 0.0734] Synergy N2 Ray 9 0.37 0.3530 [0.1568 ; 0.7949] Synergy N3 Ray 9 0.42 0.2049 [0.0444 ; 0.9469] Synergy N1 Ray 8 0.51 0.0531 [0.0216 ; 0.1306] Synergy N2 Ray 10 0.64 0.2341 [
  • Table 23 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on HT-1197 cells.
  • Table 23 Experiment Compound 1 Compound 5 Relative potency N1 (IC40) 8.91E+02 [3 59E+02 ; 2.21E+03] 8.31E+01 [4.05E+01 ; 1.70E+02] 10.7 N2 (IC40) 6.13E+02 [2.56E+02 ; 1.47E+03] 1.03E+00 [4.20E+00 ; 2.50E+01] 59.5 N3 (IC40) 7.30E+02 [4.53E+02 ; 1.18E+03] 1.61E+01 [9.15E+00 ; 2.82E+01] 45.34 N4 (IC40) 6.02E+02 [3.74E+02 ; 9.68E+02] 1.40E+01 [7.93E+00 ; 2.48E+01] 43
  • Table 24 shows interaction characteristics of Compounds (1) and (5) in HT-1197 cells.
  • Table 24 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 9 0.05 2.0559 [0.5657 ; 7.4714] Additivity N3 Ray 9 0.07 0.2116 [0.0829 ; 0.5403] synergy N4 Ray 9 0.07 0.2840 [0.1189 ; 0.6783] synergy N1 Ray 8 0.08 0.4962 [0.1566 ; 1.5728] Additivity N2 Ray 10 0.14 0.7021 [0.2170 ; 2.2724] Additivity N3 Ray 10 0.18 0.1550 [0.0708 ; 0.3392] synergy N4 Ray 10 0.19 0.1819 [0.0832 ; 0.3980] synergy N1 Ray 9 0.24 0.4944 [0.1796 ; 1.3613] Additivity N2 Ray 11 0.36 0.4624 [0.1758 ; 1.2159] Additivity N3 Ray 11 0.42 0.0947 [0.0
  • FIG. 10 shows an isobologram of growth inhibition of HT-1197 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 10A ) or Compound (5) ( FIG. 10B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) or Compound (5) showed a synergistic effect on HT-1197 cells.
  • Compound (1) shows synergy with Compound (4) and a trend to synergy with Compound (5) in KU-19-19 cells.
  • Table 25 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on KU-19-19 cells.
  • Table 25 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 2.14E+02 [1,66E202 ; 2.077E+02] 4.34E+01 [3.38E+01;5.56E+01] 4.94 N2 (IC50) 1.78E+02 [1.30E+02 ; 2.44E+02] 3.30E+01 [2.57E3010E4.023E+Ol] 5.40 N3 (IC50) 2.57E+02 [2.06E+02 ; 3.19E+02] 2.27E+01 [1.90E+01 ; 2.71E+01] 11.3 N4 (IC50) 2.63E+02 [1.96E+02 ; 3.51E+02] 2.99E+01 [2.32E+01 ; 3.85E+01] 8.79
  • Table 26 shows the interaction characterization of Compounds (1) and (4) in KU-19-19 cells.
  • Table 26 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 7 0.06 2.0177 [1.1799 ; 3.4504] Antagonism N2 Ray 7 0.05 0.5005 [0.3188 ; 0.7860] Synergy N3 Ray 8 0.08 1.1001 [0.828 ; 1.4602] additivity N4 Ray 8 0.10 0.8438 [0.5576 ; 1.2771] additivity N2 Ray 8 0.16 0.5013 [0.3150 ; 0.7978] Synergy N1 Ray 8 0.17 0.6707 [0.428 ; 1.0501] Additivity N3 Ray 9 0.23 0.9098 [0.7001 ; 1.1822] additivity N4 Ray 9 0.27 0.6537 [0.4459 ; 0.9583] synergy N2 Ray 9 0.38 0.6125 [0.3899 ; 0.9622] Synergy N1 Ray 9 0.40 0.6864 [0.4628
  • Table 27 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on KU-19-19 cells.
  • Table 27 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 2.80E+02 [2.07E+02 ; 3.79E+02] 2.60E+01 [1.95E+01 ; 3.46E+01] 10.77 N2 (IC50) 3.38+02 [2.20E+02 ; 5.19E+02] 3.04E+00 [2.07E+00 ; 4.48E+00] 111.18 N3 (IC50) 2.82E+02 [2.12E+02 ; 3.76E+02] 2.42E+00 [1.98E+00 ; 2.95E+00] 116.62 N4 (IC50) 1.94E+02 [1.53E+02 ; 2.47E+02] 2.33E+00 [1.94E+00 ; 2.79E+00] 83.34
  • Table 28 shows the interaction characterization of Compounds (1) and (5) in KU-19-19 cells.
  • Table 28 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 10 0.08 1.7399 [0.7995 ; 3.7864] additivity N3 Ray 10 0.08 0.8535 [0.6111 ; 1.1920] additivity N1 Ray 8 0.09 1.0924 [0.6788 ; 1.7580] additivity N4 Ray 10 0.11 0.8316 [0.6105 ; 1.1327] additivity N3 Ray 11 0.22 0.6394 [0.4607 ; 0.8873] synergy N2 Ray 11 0.23 1.4696 [0.7745 ; 2.7885] additivity N1 Ray 9 0.24 0.8856 [0.5602 ; 1.3999] additivity N4 Ray 11 0.29 0.7753 [0.5830 ; 1.0311] additivity N3 Ray 12 0.46 0.5302 [0.3765 ; 0.7467] synergy N2 Ray 12 0.47 1.5136 [0.8200
  • FIG. 11 shows an isobologram of growth inhibition of KU-19-19 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 11A ) or Compound (5) ( FIG. 11B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) showed a synergistic effect on KU-19-19 cells and Compound (1) in combination with Compound (5) showed a trend to synergy.
  • Compound (1) shows additivity with Compound (4) and Compound (5) in a Braf mutant melanoma cell line, A375.
  • Table 29 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on A375 cells.
  • Table 29 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 3.10E+02 [2.67E+02 ; 3.60E+02] 1.01E+01 [8.64E+00 ; 1.18E+01] 30.7 N2 (IC50) 2.88E+02 [2.41E+02 ; 3.45E+02] 1.12E+01 [9.32E+00 ; 1.35E+01] 25.7
  • Table 30 shows the interaction characteristics of Compounds (1) and (4) in A375 cells.
  • Table 30 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 8 0.03 1.0838 [0.8505 ; 1.3812] Additivity N2 Ray 8 0.04 0.9608 [0.6980 ; 1.3228] Additivity N1 Ray 9 0.10 0.9830 [0.7610 ; 1.2697] Additivity N2 Ray 9 0.11 1.1334 [0.8346 ; 1.5392] Additivity N1 Ray 10 0.25 1.0991 [0.8656 ; 1.3957] Additivity N2 Ray 10 0.28 1.1329 [0.8649 ; 1.4839] Additivity N1 Ray 11 0.52 1.1522 [0.9368 ; 1.4171] Additivity N2 Ray 11 0.56 1.028 [0.7835 ; 1.3487] Additivity N1 Ray 12 0.76 0.9289 [0.7305 ; 1.1811] Additivity N2 Ray 12 0.80 1.2189 [0.9297 ; 1.598
  • Table 31 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on A375 cells.
  • Table 31 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 2.64E+02 [2.38E+02 ; 2.92E+02] 7.01E-01 [6.32E-01 ; 7.77E-01] 376.3 N2 (IC50) 2.89E+02 [2.41E+02 ; 3.47E+02] 9.26E-01 [7.81E-01 ; 1.10E+00] 312.3
  • Table 32 shows the interaction characteristics of Compounds (1) and (5) in A375 cells.
  • Table 32 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 10 0.03 1.8132 [1.3613 ; 2.4153] Antagonism N1 Ray 11 0.08 1.1294 [0.9616 ; 1.3265] Additivity N2 Ray 11 0.10 0.8676 [0.6326 ; 1.1899] Additivity N1 Ray 12 0.21 1.2616 [1.0814 ; 1.4717] Antagonism N2 Ray 12 0.24 1.1801 [0.9184 ; 1.5164] Additivity N1 Ray 13 0.47 1.1666 [1.0061 ; 1.3528] Antagonism N2 Ray 13 0.52 1.1023 [0.8396 ; 1.4471] Additivity N1 Ray 14 0.73 1.2623 [1.0831 ; 1.4711] Antagonism N2 Ray 14 0.76 1.1830 [0.9087 ; 1.5399] Additivity N1 Ray 15 0.90 1.0731 [0.9103
  • FIG. 12 shows an isobologram of growth inhibition of A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 12A ) or with Compound (5) ( FIG. 12B ) compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) or (5) showed an additive effect on A375 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in Braf mutant melanoma cells, C32.
  • Table 33 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on C32 cells.
  • Table 33 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 8.13E+02 [5.86E+02 ; 1.13E+03] 1.74E+01 [1.47E+01 ; 2.06E+01] 46.7 N2 (IC50) 9.62E+02 [6.75E+02 ; 1.37E+03] 2.36E+01 [2.03E+01 ; 2.74E+01] 40.8 N3 (IC50) 1.45E+03 [1.19E+03 ; 1.77E+03] 2.27E+01 [2.03E+01 ; 2.54E+01] 64.1
  • Table 34 shows the interaction characteristics of Compounds (1) and (4) in C32 cells.
  • Table 34 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N3 Ray 9 0.05 0.7614 [0.6225 ; 0.9313] Synergy N1 Ray 9 0.07 0.7740 [0.5815 ; 1.0303] Additivity N2 Ray 9 0.08 0.8382 [0.6282 ; 1.1183] Additivity N3 Ray 10 0.14 0.6935 [0.5733 ; 0.8388] Synergy N1 Ray 10 0.18 0.7019 [0.5210 ; 0.9457] Synergy N2 Ray 10 0.20 0.6164 [0.4593 ; 0.8272] Synergy N3 Ray 11 0.34 0.5301 [0.4268 ; 0.6585] Synergy N1 Ray 11 0.42 0.6887 [0.4903 ; 0.9673] Synergy N2 Ray 11 0.45 0.5207 [0.3702 ; 0.7324] Synergy N3 Ray 12 0.61 0.5798 [
  • Table 35 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on C32 cells.
  • Table 35 Experiment Compound 1 Compound 5 Relative potency N1 (IC40) 8.23E+02 [4.42E+02 ; 1.53E+03] 1.13E+00 [9.89E-01 ; 1.29E+00] 727 N2 (IC40) 6.18E+02 [3.44E+02 ; 1.11E+03] 1.19E+00 [9.79E-01 ; 1.45E+00] 518.5 N3 (IC40) 2.15E+03 [1.77E+03 ; 2.62E+03] 8.46E-01 [7.06E-01 ; 1.02E+00] 2545.0 N4 (IC40) 1.07E+03 [7.24E+02 ; 1.57E+03] 7.88E-01 [5.93E-01 ; 1.05E+00] 1353.5
  • Table 36 shows the interaction characteristics of Compounds (1) and (5) in C32 cells.
  • Table 36 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N4 Ray 11 0.02 0.7309 [0.4502 ; 1.1867] Additivity N1 Ray 11 0.04 0.6662 [0.5318 ; 0.8347] Synergy N3 Ray 12 0.04 0.9525 [0.7258 ; 1.2500] Additivity N2 Ray 11 0.06 0.7353 [0.5151 ; 1.0497] Additivity N4 Ray 12 0.07 0.5630 [0.3466 ; 0.9144] Synergy N1 Ray 12 0.12 0.8010 [0.6482 ; 0.9899] Synergy N3 Ray 13 0.12 0.6808 [0.5177 ; 0.8954] Synergy N2 Ray 12 0.16 0.7696 [0.5376 ; 1.1018] Additivity N4 Ray 13 0.20 0.6761 [0.407 ; 1.1206] Additivity N3 Ray 14 0.28 0.5832 [0.4382
  • FIG. 13 shows an isobologram of growth inhibition of C32 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 13A ) or with Compound (5) ( FIG. 13B ) compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) or Compound (5) showed a synergistic effect on C32 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in SK-MEL-147 cells.
  • Table 37 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on SK-MEL-147 cells.
  • Table 37 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 5.56E+02 [3.97E+02 ; 7.77E+02] 9.12E+01 [6.45E+01 ; 1.29E+02] 6.1 N2 (IC50) 8.78E+02 [6.56E+02; 1.17E+03] 8.24E+01 [5.99E+01 ; 1.13E+02] 10.7 N3 (IC50) 8.66E+02 [6.57E+02; 1.14E+03] 8.14E+01 [6.08E+01 ; 1.09E+02] 10.6
  • Table 38 shows the interaction characteristics of Compounds (1) and (4) in SK-MEL-147 cells.
  • Table 38 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 7 0.03 0.8443 [0.4859 ; 1.4671] Additivity N1 Ray 7 0.05 0.6337 [0.3663 ; 1.0965] Additivity N2 Ray 8 0.09 0.6180 [0.3662 ; 1.0430] Additivity N3 Ray 8 0.09 0.6558 [0.3798 ; 1.1323] Additivity N1 Ray 8 0.14 0.3892 [0.228 ; 0.6636] Synergy N2 Ray 9 0.24 0.6069 [0.3901 ; 0.9443] Synergy N3 Ray 9 0.24 0.2878 [0.1692 ; 0.4896] Synergy N1 Ray 9 0.35 0.4817 [0.2814 ; 0.8248] Synergy N2 Ray 10 0.48 0.7068 [0.4690 ; 1.0652] Additivity N3 Ray 10 0.48 0.
  • Table 39 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on SK-MEL-147 cells.
  • Table 39 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 9.78E+02 [7.39E+02 ; 1.29E+03] 6.94E+00 [5.39E+00 ; 8.93E+00] 141.0 N2 (IC50) 9.68E+02 [7.93E+02 ; 1.18E+03] 6.94E+00 [5.62E+00 ; 8.57E+] 139.4 N3 (IC50) 1.08E+03 [8.80E+02 ; 1.32E+03] 7.42E+00 [6.05E+00 ; 9.10E+00] 145.2
  • Table 40 shows the interaction characteristics of Compounds (1) and (5) in SK-MEL-147 cells.
  • Table 40 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N3 Ray 10 0.06 0.6569 [0.4573 ; 0.9436] Synergy N1 Ray 10 0.07 0.6580 [0.4227 ; 1.0243] Additivity N2 Ray 10 0.07 0.8604 [0.6040 ; 1.2258] Additivity N1 Ray 11 0.19 0.5333 [0.3346 ; 0.8498] Synergy N2 Ray 11 0.19 0.5885 [0.4167 ; 0.8311] Synergy N3 Ray 11 0.19 0.5846 [0.4240 ; 0.8061] Synergy N1 Ray 12 0.41 0.6460 [0.4447 ; 0.9383] Synergy N3 Ray 12 0.41 0.4436 [0.3235 ; 0.6084] Synergy N2 Ray 12 0.42 0.5921 [0.4299 ; 0.8156] Synergy N1 Ray 13 0.70
  • FIG. 14 shows an isobologram of growth inhibition of SK-MEL-147 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 14A ) or Compound (5) ( FIG. 14B ), compared to additive growth inhibition.
  • the combination of Compound (1) and Compound (4) or (5) showed a synergistic effect on SK-MEL-147 cells.
  • Compound (1) shows additivity with Compound (4) in DV90 cells.
  • Table 41 shows the IC50 estimations for each compound used alone and relative potency of Compounds (1) and (4) on DV90 cells.
  • Table 41 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 1.40E+02 [6.90E+01 ; 2.85E+02] 3.64E+01 [2.10E+01 ; 6.30E+01] 3.9 N2 (IC50) 1.63E+02 [9.22E+01 ; 2.87E+02] 1.97E+01 [9.77E+00 ; 3.99E+01] 8.2 N3 (IC50) 2.54E+02 [1.48E+02 ; 4.35E+02] 5.55E+01 [3.23E+01 ; 9.52E+01] 4.6
  • Table 42 shows the interaction characteristics of Compounds (1) and (4) in DV90 cells.
  • Table 42 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 6 0.03 1.1814 [0.428 ; 3.2561] Additivity N2 Ray 7 0.04 1.8696 [0.5511 ; 6.3424] Additivity N3 Ray 7 0.06 0.4777 [0.1699 ; 1.3435] Additivity N1 Ray 7 0.07 0.7219 [0.2082 ; 2.5032] Additivity N2 Ray 8 0.11 0.9143 [0.31999; 2.6131] Additivity N3 Ray 8 0.18 1.4945 [0.5946 ; 3.7567] Additivity N1 Ray 8 0.21 0.7855 [0.2519 ; 2.4499] Additivity N2 Ray 9 0.29 1.0474 [0.3818 ; 2.8733] Additivity N3 Ray 9 0.42 0.6976 [0.2907 ; 1.6745] Additivity N1 Ray 9 0.46 0.8797 [0.3443 ; 2.
  • Table 43 shows the IC50 estimations for each compound used alone and relative potency of Compounds (1) and (5) on DV90 cells.
  • Table 43 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 1.39E+02 [8.65E+01 ; 2.23E+02] 3.23E+00 [2.20E+00 ; 4.72E+00] 43.1 N2 (IC50) 7.83E+01 [3.74E+01 ; 1.64E+02] 1.20E+00 [6.47E-01 ; 2.24E+00] 65.0 N3 (IC50) 2.07E+02 [1.57E+02 ; 2.72E+02] 9.03E-01 [4.62E-01 ; 1.77E+00] 228.9
  • Table 44 shows the interaction characteristics of Compounds (1) and (5) in DV90 cells.
  • Table 44 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N3 Ray 10 0.04 3.2416 [1.5428 ; 6.8111] Antagonism N2 Ray 9 0.05 0.4163 [0.1210 ; 1.4320] Additivity N1 Ray 9 0.07 1.6379 [0.8013 ; 3.3482] Additivity N2 Ray 10 0.13 0.8221 [0.3180 ; 2.1249] Additivity N3 Ray 11 0.13 2.6946 [1.3277 ; 5.4687] Antagonism N1 Ray 10 0.19 0.8337 [0.4408 ; 1.5766] Additivity N3 Ray 12 0.30 1.6455 [0.9386 ; 2.8849] Additivity N2 Ray 11 0.34 0.1919 [0.0804 ; 0.4583] Synergy N1 Ray 11 0.44 0.6070 [0.3066 ; 1.2017] Additivity N3 Ray 13 0.59 1.0816 [0.6800 ;
  • FIG. 15 shows an isobologram of growth inhibition of DV90 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 15A ) or Compound (5) ( FIG. 15B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) showed additivity on DV90 cells.
  • Compound (1) shows synergy with Compound (5) in H460 cells.
  • Table 45 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on H460 cells.
  • Table 45 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 3.19E+02 [2.61E+02 ; 3.89E+02] 1.55E+02 [1.07E+02 ; 2.24E+02] 2.1 N2 (IC50) 1.66E+02 [1.25E+02 ; 2.20E+02] 8.40E+01 [6.20E+01 ; 1.14E+02] 2.0 N3 (IC50) 1.36E+02 [9.00E+01 ; 2.07E+02] 1.04E+02 [6.48E+01 ; 1.68E+02] 1.3
  • Table 46 shows the interaction characteristics of Compounds (1) and (4) in H460 cells.
  • Table 46 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 6 0.05 1.5331 [0.9970 ; 2.3574] Additivity N2 Ray 6 0.05 0.7694 [0.3938 ; 1.5031] Additivity N3 Ray 6 0.07 0.7949 [0.3405 ; 1.8555] Additivity N1 Ray 7 0.13 0.7955 [0.4571 ; 1.3843] Additivity N2 Ray 7 0.13 1.1173 [0.6732 ; 1.8545] Additivity N3 Ray 7 0.19 0.7763 [0.3564 ; 1.6910] Additivity N1 Ray 8 0.33 0.5376 [0.3261 ; 0.8862] Synergy N2 Ray 8 0.34 1.3456 [0.8795 ; 2.0587] Additivity N3 Ray 8 0.43 1.7012 [0.8964 ; 3.2285] Additivity N1 Ray 9 0.62 0.6094 [0.3864
  • Table 47 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on H460 cells.
  • Table 47 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 2.53E+02 [2.15E+02 ; 2.99E+02] 6.82E+00 [4.95E+00 ; 9.39E+00] 37.1 N2 (IC50) 3.19E+02 [2.72E+02 ; 3.75E+02] 1.01E+01 [7.69E+00 ; 1.34E+01] 31.5 N3 (IC50) 2.97E+02 [2.29E+02 ; 3.87E+02] 1.10E+01 [7.67E+00 ; 1.59E+01] 26.9
  • Table 48 shows the interaction characteristics of Compounds (1) and (5) in H460 cells.
  • Table 48 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N3 Ray 8 0.04 1.7372 [0.9459 ; 3.1903] Additivity N2 Ray 9 0.10 0.7189 [0.4787 ; 1.0795] Additivity N3 Ray 9 0.11 0.7200 [0.4072 ; 1.2730] Additivity N1 Ray 10 0.21 0.7177 [0.4766 ; 1.0808] Additivity N2 Ray 10 0.24 0.7328 [0.5205 ; 1.0318] Additivity N3 Ray 10 0.27 0.9753 [0.6162 ; 1.5438] Additivity N1 Ray 11 0.47 0.6777 [0.4884 ; 0.9405] Synergy N2 Ray 11 0.51 0.6608 [0.4894 ; 0.8923] Synergy N3 Ray 11 0.55 0.2378 [0.1355 ; 0.4172] Synergy N1 Ray 12 0.73 0.6800 [0.504
  • FIG. 16 shows an isobologram of growth inhibition of H460 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 16A ) or Compound (5) ( FIG. 16B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (5) showed a synergistic effect on H460 cells.
  • Compound (1) shows additivity with Compounds (4) and (5) in UM-UC-3 cells.
  • Table 49 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on UM-UC-3 cells.
  • Table 49 Experiment Compound 1 Compound 4 Relative potency N1 (IC50) 8.53E+03 [6.54E+03 ; 1.11E+04] 2.63E+02 [1.53E+02 ; 4.52E+02] 32.48 N2 (IC50) 6.00E+03 [4.86E+03 ; 7.41E+03] 7.37E+01 [4.92E+01 ; 1.10E+02] 81.43
  • Table 50 shows the interaction characteristics of Compounds (1) and (4) in UM-UC-3 cells.
  • Table 50 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N2 Ray 9 0.04 1.7681 [0.9837; 3.1780] Additivity N1 Ray 9 0.09 1.2061 [0.5751; 2.5293] Additivity N2 Ray 10 0.11 1.0148 [0.5706; 1.8047] Additivity N1 Ray 10 0.24 0.7708 [0.4036; 1.4723] Additivity N2 Ray 11 0.29 0.6674 [0.3921; 1.1360] Additivity N1 Ray 11 0.51 0.6154 [0.3304; 1.1462] Additivity N2 Ray 12 0.55 0.9561 [0.7191; 1.2711] Additivity N1 Ray 12 0.75 0.7632 [0.4881; 1.1933] Additivity N2 Ray 13 0.80 0.8798 [0.6518; 1.1875] Additivity N1 Ray 13 0.91 0.9695 [0.6166; 1.5244] Additivity N2
  • Table 51 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on UM-UC-3 cells.
  • Table 51 Experiment Compound 1 Compound 5 Relative potency N1 (IC50) 6.95E+03 [5.65E+03 ; 8.55E+03] 2.24E+02 [1.31E+02 ; 3.85E+02] 31.0 N2 (IC50) 5.40E+03 [4 . 54E+03 ; 6.43E+03] 2.34E+01 [1.65E+01 ; 3.33E+01] 230.5 N3 (IC50) 5.75E+03 [4.52E+03 ; 7.31E+03] 2.30E+01 [1.43E+01 ; 3.70E+01] 249.7
  • Table 52 shows the interaction characteristics of Compounds (1) and (5) in UM-UC-3 cells.
  • Table 52 Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization N1 Ray 8 0.03 1.0778 [0.4454 ; 2.6084] Additivity N2 Ray 10 0.04 0.9437 [0.5595 ; 1.5917] Additivity N3 Ray 10 0.04 1.1040 [0.5600 ; 2.1765] Additivity N1 Ray 9 0.10 0.9785 [0.4360 ; 2.1962] Additivity N3 Ray 11 0.12 0.8211 [0.4481 ; 1.5044] Additivity N2 Ray 11 0.13 0.8543 [0.5354 ; 1.3630] Additivity N1 Ray 10 0.24 1.4840 [0.6757 ; 3.2592] Additivity N3 Ray 12 0.29 0.6503 [0.3812 ; 1.1094] Additivity N2 Ray 12 0.30 0.8290 [0.5545 ; 1.2395] Additivity N1 Ray 11 0.52 0.8182 [0.4922
  • FIG. 17 shows an isobologram of growth inhibition of UM-UC-3 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) ( FIG. 17A ) or Compound (5) ( FIG. 17B ), compared to additive growth inhibition.
  • Compound (1) in combination with Compound (4) or Compound (5) showed additivity on UM-UC-3 cells.

Abstract

Methods of causing growth inhibition of cancer cells are provided, wherein the methods comprise contacting cancer cells with an HDM2 antagonist and a MEK inhibitor. Compositions in which the HDM2 antagonist and MEK inhibitor are combined also are provided.

Description

    BACKGROUND
  • HDM2 negatively regulates p53 activity by binding to the transactivation domain of p53 and by acting as a p53-specific E3 ubiquitin ligase. In response to diverse stresses, p53 can induce cell cycle arrest and apoptosis.
  • Compound (1)
    Figure imgb0001
    is a selective antagonist of HDM2. ( WO2012/065022 ). Binding of Compound (1) to HDM2 disrupts the interaction between HDM2 and p53 thereby inhibiting the ability of HDM2 to bind to and inhibit p53.
  • Compound (4), N-((S)-2,3-dihydroxypropyl)-3-(2-fluoro-4-iodo-phenylamino)isonicotinamide (also referred to as Pimasertib, MSC1936369, or AS703026) is a novel, allosteric inhibitor of MEK. It possesses relatively high potency and selectivity, having no activity against 217 kinases or 90 non-kinase targets when tested at 10 µM. ( WO06/045514 ).
  • Tumor cells treated with inhibitors of MEK kinases typically respond via inhibition of phosphorylation of ERK, down-regulation of Cyclin D, induction of G1 arrest, and finally undergoing apoptosis. The effect of MEK inhibition on tumor growth in selected mutant xenograft tumors that express mutant B-raf and selected Ras mutant tumors are described in D. B. Solit et al., Nature 2006; 439: 358-362.
  • Predominant toxicities reported for MEK inhibitors in human patients include diarrhea, fatigue, rash, vomiting, nausea, and reversible visual disturbances. (Haura et al., Clin Cancer Res (2010) 16(8):2450-2457). In particular, similar to epidermal growth factor receptor inhibitors, dermatologic toxicities such as papulopustular rash, xerosis, and pruritus are associated with MEK inhibitors. (Baladula et al., Invest New Drugs (2011) 29:1114-1121).
  • There remains a need for a cancer therapy that is more effective in inhibiting cell proliferation and tumor growth while minimizing patient toxicity. There is a particular need for an HDM2 antagonist and/or MEK inhibitor therapy that is made more efficacious without substantially increasing, or even maintaining or decreasing, the dosages of HDM2 antagonist and/or MEK inhibitor traditionally used in the art.
  • SUMMARY
  • Provided herein are compositions for the treatment of patients having cancer and methods of using the compositions that employ an HDM2 antagonist in combination with an inhibitor of MEK pathway signaling.
  • In a first aspect, methods for treating a subject having cancer are provided comprising administering to the subject an effective amount of Compound (1):
    Figure imgb0002
    or a pharmaceutically acceptable salt thereof and Compound (4)
    Figure imgb0003
    or a pharmaceutically acceptable salt thereof. In some embodiments, Compound (1) and/or Compound (4) are administered with a pharmaceutically acceptable carrier. In some embodiments, Compound (1) and Compound (4) are administered in either separate or single dosage forms. In some embodiments, Compound (1) and Compound (4) are administered at different times. In some embodiments, Compound (1) and Compound (4) are administered at substantially the same time. In some embodiments, the effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  • In a second aspect, a combination is provided for use in treating a subject having cancer, the combination comprising a therapeutically effective amount of Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof. In one embodiment, the combination is in separate dosage forms, while in another embodiment, the combination is in a single dosage form. In a third aspect, a use of a combination comprising a therapeutically effective amount of Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in treatment of lymphoma is provided. In one embodiment, the combination is in separate dosage forms, while in another embodiment, the combination is in a single dosage form.
  • In some embodiments of the second and third aspect, the therapeutically effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  • In a fourth aspect, a composition comprising Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof is provided. The composition can further comprise a pharmaceutically acceptable carrier.
  • In a fifth aspect, kits are provided comprising: (A) Compound (1), or a pharmaceutically acceptable salt thereof; (B) Compound (4), or a pharmaceutically acceptable salt thereof; and (C) instructions for use. In some embodiments, the instructions are for the use of Compounds (1) and (4) in the treatment of cancer.
  • In some embodiments of aspects four and five, a therapeutically effective amount of Compound (1) and Compound (4) provided. In some embodiments, a therapeutically effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  • In some embodiments of aspects one through five, the cancer expresses mutant Nras. In one example, the mutant Nras expressing cancer is melanoma. In another example, the Nras mutant expressing cancer is bladder cancer. In other embodiments, the cancer expresses mutant Braf. In one example, the Braf mutant expressing cancer is melanoma.
  • BRIEF DESCRIPTION OF THE DRAWINGS
    • FIG. 1 shows a table of the mean synergy score and False Discovery Rate (FDR) for HDM2 antagonist Compounds (1), (2), and (3) in combination with MEK inhibitor Compounds (6), (7), (8), (9), and (10), in the indicated cell lines (A: 769-P, B: A375, C: BPH1, D:D32, E: Cake-1, F: DBTRG-05MG, G: DK-MG, .H: H4, I: HCT-116, J: HT-1197, K: JAR, L: LNCaP-sa, M: MCF7, N: MG-63, O: MV-4-11, P: RKO, Q: SJSA-1, R: SK-N-AS, S: SR, and T: SW48).
    • FIG. 2 shows: A) percent growth inhibition of C32 cells when treated with the indicated amounts of Compound (1) in combination with Compound (6); B) the level of growth inhibition of C32 cells versus drug concentration where ◆ is Compound (1), ■ is Compound (6), and ▲ is Compound (1) in combination with Compound (6).
    • FIG 3 shows: A) percent growth inhibition of MV4-11 cells when treated with the indicated amounts of Compound (1) in combination with Compound (4), and B) the level of growth inhibition of MV4-11 cells versus drug concentration where ◆ is Compound (1), ■ is Compound (6), and ▲ is Compound (1) in combination with Compound (6).
    • FIG. 4 shows: A) percent growth inhibition of A375 cells when treated with the indicated amounts of Compound (1) in combination with Compound (6), and B) an isobologram of growth inhibition of A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (6) compared to additive growth inhibition.
    • FIG. 5 shows: A) percent growth inhibition of Vemurafenib resistant A375 cells when treated with the indicated amounts of Compound (1) in combination with Compound (6), and B) an isobologram of growth inhibition of Vemurafenib resistant A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (6) compared to additive growth inhibition.
    • FIG. 6 shows an isobologram of growth inhibition of C8161 cells treated with the Compound (1) in combination with Compound (4) (A) or with Compound (5) compared to additive growth inhibition where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles and triangles represent a first and second experiment, respectively.
    • FIG. 7 shows an isobologram of growth inhibition ofHCT116 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, squares, and astrices represent experiments 1-4, respectively.
    • FIG. 8 shows an isobologram of growth inhibition of SK-MEL-103 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, and squares represent experiments 1-3, respectively.
    • FIG. 9 shows an isobologram of growth inhibition of UACC-62 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, and squares represent experiments 1-3, respectively.
    • FIG. 10 shows an isobologram of growth inhibition ofHT-1197 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC40 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC40 of Compound (1); triangles, squares, and astrices represent experiments 1-3, respectively.
    • FIG. 11 shows an isobologram of growth inhibition of KU-19-19 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, squares, and astrices represent experiment 1-4, respectively.
    • FIG. 12 shows an isobologram of growth inhibition of A375 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles and triangles represent experiments 1 and 2, respectively.
    • FIG. 13 shows an isobologram of growth inhibition C32 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, and squares represent experiments 1-3, respectively.
    • FIG. 14 shows an isobologram of growth inhibition SK-MEL-147 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, and squares represent experiments 1-3, respectively.
    • FIG. 15 shows an isobologram of growth inhibition DV90 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, and squares represent experiments 1-3, respectively.
    • FIG. 16 shows an isobologram of growth inhibition H460 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles, triangles, and squares represent experiments 1-3, respectively.
    • FIG. 17 shows an isobologram of growth inhibition UM-UC-3 cells treated with Compound (1) in combination with Compound (4) (A) or with Compound (5) (B) where the y-axis is [Compound (4) or (5)]/IC50 of Compound (4) or (5), respectively and the x-axis is [Compound (1)]/IC50 of Compound (1); circles and triangles represent experiments 1 and 2, respectively.
    DETAILED DESCRIPTION
  • Compositions and methods of use thereof for treatment of Nras mutant expressing bladder cancer or melanoma and for the treatment of mutant Braf expressing melanoma are provided herein that employ an HMD2 antagonist in combination with a MEK inhibitor. The compositions provided herein afford better efficacy and/or potency than the single agent components of the compositions. As shown herein for the first time, Compound (1) demonstrates synergistic activity in combination with Compound (4) in causing growth inhibition of Nras mutant expressing bladder cancer cells, mutant Nras expressing melanoma cells, and mutant Braf expressing melanoma cells.
  • Methods for treating a subject having cancer such as Nras mutant expressing bladder cancer or melanoma and for the treatment of mutant Braf expressing melanoma are provided. In one embodiment, the methods comprise administering to the patient a therapeutically effective amount of an HDM2 antagonist and MEK inhibitor as further described herein.
  • In some embodiments, the methods and compositions comprise a HDM2 antagonist having the formula (1):
    Figure imgb0004
  • The compound according to formula (1) is referred to herein as "Compound (1)". The preparation and properties of Compound (1) are provided in, for example, International Patent Publication No. WO2012/065022 , particularly Example 11, p. 169-171 therein. The entire contents of WO2012/065022 are incorporated herein by reference.
  • In some embodiments, the methods and compositions comprise a MEK inhibitor having the formula (4):
    Figure imgb0005
  • The compound according to formula (4) is referred to herein as "Compound (4)". The preparation and properties of Compound (4), also referred to as Pimasertib, are provided in, for example, International Patent Publication No. WO06/045514 , particularly Example 115 and Table 1 therein. The entire contents of WO06/045514 are incorporated herein by reference.
  • In some embodiments, the compounds described herein are unsolvated. In other embodiments, one or both of the compounds used are in solvated form. As known in the art, the solvate can be any ofpharmaceutically acceptable solvent, such as water, ethanol, and the like. In general, the presence of a solvate or lack thereof does not have a substantial effect on the efficacy of Compounds (1) or (4) described above. Although the Compounds (1) and (4) are depicted in their neutral forms, in some embodiments, these compounds are used in a pharmaceutically acceptable salt form. The salt can be obtained by any of the methods well known in the art. A "pharmaceutically acceptable salt" of the compound refers to a salt that is pharmaceutically acceptable and that retains pharmacological activity. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, or S. M. Berge, et al., "Pharmaceutical Salts," J. Pharm. Sci., 1977; 66:1-19, both of which are incorporated herein by reference.
  • Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, as well as those salts formed with organic acids, such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, p-toluenesulfonic acid, and salicylic acid.
  • In some embodiments of the methods and uses provided herein, Compound (1) is administered simultaneously with Compound (4). Simultaneous administration typically means that both compounds enter the patient at precisely the same time. However, simultaneous administration also includes the possibility that Compound (1) and Compound (4) enter the patient at different times, but the difference in time is sufficiently miniscule that the first administered compound is not provided the time to take effect on the patient before entry of the second administered compound. Such delayed times typically correspond to less than 1 minute, and more typically, less than 30 seconds.
  • In one example, wherein the compounds are in solution, simultaneous administration can be achieved by administering a solution containing the combination of compounds. In another example, simultaneous administration of separate solutions, one of which contains the Compound (1) and the other of which contains Compound (4), can be employed. In one example wherein the compounds are in solid form, simultaneous administration can be achieved by administering a composition containing the combination of compounds.
  • In other embodiments, Compound (1) and Compound (4) are not simultaneously administered. In one set of embodiments, the Compound (1) is administered before Compound (4). In another set of embodiments, Compound (4) is administered before Compound (1). The time difference in non-simultaneous administrations can be greater than 1 minute, and can be, for example, precisely, at least, up to, or less than 5 minutes, 10 minutes, 15 minutes, 30 minutes, 45 minutes, 60 minutes, two hours, three hours, six hours, nine hours, 12 hours, 24 hours, 36 hours, or 48 hours. In another embodiment, the first administered compound is provided time to take effect on the patient before the second administered compound is administered. Generally, the difference in time does not extend beyond the time for the first administered compound to complete its effect in the patient, or beyond the time the first administered compound is completely or substantially eliminated or deactivated in the patient.
  • In some embodiments, one or both of the Compounds (1) and (4) are administered in a therapeutically effective (i.e., therapeutic) amount or dosage. A "therapeutically effective amount" is an amount of Compound (1) or Compound (4) that, when administered to a patient by itself, effectively treats the cancer (for example, inhibits tumor growth, stops tumor growth, or causes tumor regression). An amount that proves "therapeutically effective amount" in a given instance, for a particular subject, may not be effective for 100% of subjects similarly treated for the disease or condition under consideration, even though such dosage is deemed a "therapeutically effective amount" by skilled practitioners. The amount of the compound that corresponds to a therapeutically effective amount is affected by the type of cancer, stage of the cancer, the age of the patient being treated, and other facts.
  • In other embodiments, one or both of the Compounds (1) and (4) are administered in a sub-therapeutically effective amount or dosage. A sub-therapeutically effective amount is an amount of Compound (1) or (4) that, when administered to a patient by itself, does not completely inhibit over time the biological activity of the intended target.
  • Whether administered in therapeutic or sub-therapeutic amounts, the combination of Compounds (1) and (4) should be effective in treating, for example mutant Nras expressing melanoma mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma. For example, a sub-therapeutic amount of Compound (4) can be an effective amount if, when combined with Compound (1), the combination is effective in the treatment of mutant Nras expressing melanoma mutant Nras expressing bladder cancer, or mutant Braf expressing melanoma.
  • The combination of compounds provided herein exhibits a synergistic effect (i.e., greater than additive effect) in the treatment of the cancer. The term "synergistic effect" as used herein, refers to action of two agents such as, for example, Compound (1) and Compound (4), producing an effect, for example, slowing the symptomatic progression of cancer or symptoms thereof, which is greater than the simple addition of the effects of each drug administered by themselves. A synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emax equation (Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet. 6: 429-453 (1981)), the equation of Loewe additivity (Loewe, S. and Muischnek, H., Arch. Exp. Pathol Pharmacol. 114: 313-326 (1926)) and the median-effect equation (Chou, T. C. and Talalay, P., Adv. Enzyme Regul. 22: 27-55 (1984)). Each equation referred to above can be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. The corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively.
  • In different embodiments, depending on the combination and the effective amounts used, the combination of compounds can inhibit cancer growth. In some embodiments, the combination achieves cancer stasis. In some embodiments, the combination achieves substantial or complete cancer regression.
  • As used herein, the term "about" generally indicates a possible variation of no more than 10%, 5%, or 1% of a value. For example, "about 25 mg/kg" will generally indicate, in its broadest sense, a value of 22.5-27.5 mg/kg, i.e., 25 ± 10 mg/kg.
  • While the amounts of Compounds (1) and (4) should result in the effective treatment of a cancer, the amounts, when combined, are preferably not excessively toxic to the patient (i.e., the amounts are preferably within toxicity limits as established by medical guidelines). In some embodiments, either to prevent excessive toxicity and/or provide a more efficacious treatment of the cancer, a limitation on the total administered dosage is provided. Typically, the amounts considered herein are per day; however, half-day and two-day or three-day cycles also are considered herein.
  • Different dosage regimens may be used to treat the cancer. In some embodiments, a daily dosage, such as any of the exemplary dosages described above, is administered once, twice, three times, or four times a day for three, four, five, six, seven, eight, nine, or ten days. Depending on the stage and severity of the cancer, a shorter treatment time (e.g., up to five days) may be employed along with a high dosage, or a longer treatment time (e.g., ten or more days, or weeks, or a month, or longer) may be employed along with a low dosage. In some embodiments, a once- or twice-daily dosage is administered every other day. In some embodiments, each dosage contains both Compound (1) and Compound (4) to be delivered as a single dosage, while in other embodiments, each dosage contains either Compound (1) or Compound (4) to be delivered as separate dosages.
  • Examples of types of cancers to be treated with the present invention include, for example, mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  • The subject considered herein is typically a human, for example a human patient. However, the subject can be any mammal for which cancer treatment is desired. Thus, the methods described herein can be applied to both human and veterinary applications.
  • The term "treating" or "treatment", as used herein, indicates that the method has, at the least, mitigated abnormal cellular proliferation. For example, the method can reduce the rate of cancer growth in a patient, or prevent the continued growth of a cancer, or even reduce the overall reach of the cancer
  • Compounds (1) and (4), or their pharmaceutically acceptable salts or solvate forms, in pure form or in an appropriate pharmaceutical composition, can be administered via any of the accepted modes of administration or agents known in the art. The compounds can be administered, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally. The dosage form can be, for example, a solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, pills, soft elastic or hard gelatin capsules, powders, solutions, suspensions, suppositories, aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages. A particular route of administration is oral, particularly one in which a convenient daily dosage regimen can be adjusted according to the degree of severity of the disease to be treated.
  • In one aspect, a composition is provided that comprises Compound (1) and Compound (4). In some embodiments, the composition is in the form of a solid (e.g., a powder or tablet) including Compound (1) and Compound (4) in solid form, and optionally, one or more auxiliary (e.g., adjuvant) or pharmaceutically active compounds in solid form. In other embodiments, the composition further includes any one or combination of pharmaceutically acceptable carriers (i.e., vehicles or excipients) known in the art, thereby providing a liquid dosage form.
  • As discussed above, Compound (1) and Compound (4) of the combination therapy can be administered in a single unit does or separate dosage forms. Accordingly, the pharmaceutically acceptable carriers and excipients described throughout the application can be combined with Compounds (1) and (4) in a single unit dose, as well as individually combined with Compounds (1) and (4) when these compounds are administered separately.
  • Auxiliary and adjuvant agents may include, for example, preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms is generally provided by various antibacterial and antifungal agents, such as, parabens, chlorobutanol, phenol, sorbic acid, and the like. Isotonic agents, such as sugars, sodium chloride, and the like, may also be included. Prolonged absorption of an injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. The auxiliary agents also can include wetting agents, emulsifying agents, pH buffering agents, and antioxidants, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylated hydroxytoluene, and the like.
  • Dosage forms suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, magnesium stearate and the like (h) adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agents.
  • Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well-known in the art. They can contain pacifying agents and can be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds also can be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., the HDM2 antagonist or the MEK inhibitor compound described herein, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3- butyleneglycol, dimethyl formamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
  • Suspensions, in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • Compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds described herein with, for example, suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • Dosage forms for topical administration may include, for example, ointments, powders, sprays, and inhalants. The active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as can be required. Ophthalmic formulations, eye ointments, powders, and solutions also can be employed.
  • Generally, depending on the intended mode of administration, the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of the compounds described herein, or a pharmaceutically acceptable salt thereof, and 99% to 1% by weight of a pharmaceutically acceptable excipient. In one example, the composition will be between about 5% and about 75% by weight of a compounds described herein, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art. Reference is made, for example, to Remington's Pharmaceutical Sciences, 18th Ed., (Mack Publishing Company, Easton, Pa., 1990).
  • In some embodiments, the composition includes one or more other anti-cancer compounds. For example, administered compositions can comprise standard of care agents for the type of cancers selected for treatment.
  • In another aspect, kits are provided. Such kits can comprise package(s) comprising compounds or compositions as described herein. In one embodiment, kits comprise Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4) or a pharmaceutically acceptable salt thereof.
  • The phrase "package" means any vessel containing compounds or compositions presented herein. In some embodiments, the package can be a box or wrapping. Packaging materials for use in packaging pharmaceutical products are well-known to those of skill in the art. Examples of pharmaceutical packaging materials include, but are not limited to, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • The kit also can contain items that are not contained within the package but are attached to the outside of the package, for example, pipettes.
  • Kits can contain instructions for administering compounds or compositions of the invention to a patient. Kits also can comprise instructions for approved uses of compounds herein by regulatory agencies, such as the United States Food and Drug Administration. Kits also can contain labeling or product inserts for the inventive compounds. The package(s) and/or any product insert(s) may themselves be approved by regulatory agencies. The kits can include compounds in the solid phase or in a liquid phase (such as buffers provided) in a package. The kits also can include buffers for preparing solutions for conducting the methods, and pipettes for transferring liquids from one container to another.
  • Examples have been set forth below for the purpose of illustration and to describe certain specific embodiments of the invention. However, the scope of the claims is not to be in any way limited by the examples set forth herein.
  • EXAMPLE 1 HDM2-p53 Inhibitors
  • Three HDM2-p53 inhibitors, Compounds
    Figure imgb0006
    Figure imgb0007
    were examined in combination with MEK inhibitors
    Figure imgb0008
    Figure imgb0009
    Figure imgb0010
    Figure imgb0011
    and
    Figure imgb0012
  • The preparation and properties of Compound (5), also referred to as Trametinib, GSK1120212, GSK212, and JTP-74057), are provided in, for example, in published PCT application WO2005/121142 , the contents of which are incorporated herein by reference.
  • The preparation and properties of Compound (6), also referred to as AZD6244, Selumetinib, and ARRY-142886) are provided in, for example, in published PCT application WO03/077914 , particularly Example 10 therein, the contents of which are incorporated herein by reference.
  • The preparation and properties of Compound (7), also referred as PD0325901 (N-(2,3-dihydroxy-propoxy)-3,4-difluoro-2-(2-fluoro-4-iodo-phenylamino)-benzamide) are provided in, for example, in published PCT application WO2002006213 , particularly Example 85 therein, the contents of which are incorporated herein by reference.
  • The preparation and properties of Compound (8), also referred as also referred to as 4-[2-(2-chloro-4-fluorophenylamino)-5-methylpyrimidin-4-yl]-N-[1(S)-(3-chlorophenyl)-2-hydroxyethyl]-1H-pyrrole-2-carboxamide are provided in, for example, in Aronov, et al, J.Med. Chem. (2009) 52:6362-6368, the contents of which are incorporated herein by reference.
  • The preparation and properties of Compound (9), also referred to as RO-5126766 (N-[3-fluoro-4-[4-methyl-2-oxo-7-(pyrimidin-2-yloxy)-2H-1-benzopyran-3-ylmethyl]pyridin-2-yl]-N'-methylsulfamide), are provided in, for example, in published US patent application US2010/0004233 , particularly paragraphs [1780]-[1793] therein, the contents of which are incorporated herein by reference.
  • The preparation and properties of Compound (10), also referred to as (6-methoxy-7-[3-(4-morpholinyl)propoxy]-4-(4-phenoxyphenylamino)quinoline-3-carbonitrile), are provided in, for example, in Zhang, et al, Bioor. Med. Chem. Let. (2000) 10:2825-2828, the contents of which are incorporated herein by reference.
  • The binding affinity of Compounds (1), (2), and (3) for MDM2 was determined as described in published PCT application WO2012/065022 , Examples 2 and 3, and shown herein in Table 1. Table 1
    COMPOUND IC50 (MDM2)
    (1) 8.3 ± 3.0 nM
    (2) 5.6 ± 1.7 nM
    (3) 104 nM
  • The combinations of HDM2-p53 inhibitor and MEK inhibitor were examined in a panel of twenty cell lines (Table 2). The panel of cell lines included cell lines that were induced to undergo apoptosis upon exposure to Compound (1) (apoptosis), as well as cell lines where exposure to Compound (1) resulted in inhibition of cell proliferation but not apoptosis (stasis), and cell lines that did not undergo apoptosis or experience stasis upon exposure to Compound (1) (resistant).
  • A 36-point combination dose response matrix over a treatment time of seventy-two hours was used. The method by which this screen was performed is described below. Table 2
    Cell Line Tissue Tumor Type Effect of Cpd (1) on Cells in vitro Ras pathway status
    BPH1 Prostate Hyperplasia resistant
    MG-63 Bone Osteosarcoma resistant
    SK-N-AS Adrenal Neuroblastoma resistant
    Renal Cell
    769-P Kidney Adenocarcinoma stasis
    C32 Skin Melanoma stasis Braf mut
    Caki-1 Kidney Carcinoma stasis
    H4 Brain Neuroglioma stasis
    Carcinoma,
    HCT-116 Colorectal Colorectal stasis Kras mut
    HT-1197 Bladder Carcinoma stasis Nras mut
    Analplastic large
    SR Lymphomacell lymphoma stasis
    SW48 Colorectal Adenocarcinoma stasis EGFR mut
    MCF7 Breast Adenocarcinoma stasis
    LNCaP Prostate AdenoCarcinoma stasis
    Carcinoma,
    RKO Colorectal Colorectal apoptosis Braf mut
    A375 Melanoma Melanoma apoptosis Brafmut
    DBTRG-
    05MG Brain Glioblastoma apoptosis Braf mut
    DK-MG1 Brain Glioblastoma apoptosis
    JAR1 Placenta Choriocarcinoma apoptosis
    Acute
    Myelogenous
    MV-4-11 Leukemia Leukemia apoptosis Flt3ITD
    SJSA-11 Bone Sarcoma apoptosis
    1. HDM2 amplified
  • Cell Culture
  • Cells were thawed from a liquid nitrogen preserved state. Once cells had been expanded and divided at their expected doubling times, cells were seeded in complete media (Table 3) at 100 cells per well in black 1536 tissue culture treated plates. Assay plates were centrifuged for 2 min at 335 x g, incubated at room temperature for 20 min, and incubated at 37° C for 24 hours before treatment. Table 3
    Cell Line Growth Conditions
    769-P RPMI WITH 10% FBS
    A375 DMEM, 10% FBS
    BPH1 RPMI, 20% FBS,
    C32 Eagles MEM 10% FBS
    Caki-1 McCoy's 5A, 10% FBS
    DBTRG-05MG RPMI, 10% FBS
    DK-MG1 RPMI, 10% FBS
    H4 DMEM, 10% FBS
    HCT-116 McCoy's 5A, 10% FBS
    HT-1197 Eagles MEM 10% FBS
    JAR1 RPMI, 10% FBS
    LNCaP DMEM, 10% FBS
    Eagles MEM
    10% FBS, 0.01
    MCF7 mg/ml bovine insulin
    MG-63 Eagles MEM 10% FBS
    MV-4-11 Iscoves, 10% FBS
    RKO Eagles MEM 10% FBS
    SJSA-11 RPMI, 10% FBS
    DMEM, 10% FBS, 0.1 mM
    SK-N-AS NEAA
    SR RPMI, 10% FBS
    SW48 RPMI, 10% FBS, 5% CO2
  • ATP levels were used as a measure of cell number (cell growth). At the time of treatment, ATP levels were measured in an untreated set of assay plates (Tzero plates). ATP levels were measured by adding ATPLite (Perkin Elmer) according to manufacturer's instructions. The Tzero plates were read using ultra-sensitive luminescence on Envision Plate Readers. Assay plates receiving treatment were incubated with compound for 72 h at 37° C with 5% CO2. After 72 h, plates were developed for endpoint analysis using ATPLite.
  • Assay plates were accepted for analysis if they passed the following quality control standards: relative luciferase values were consistent throughout the entire experiment, Z' factor scores were greater than 0.6, untreated/vehicle controls behaved consistently on the plate. The calculation for synergy score is provided below.
  • Calculation of Growth Inhibition
  • Growth Inhibition (GI) was used as a measure of cell viability. As described above, ATP levels were measured at the time of dosing (T0) and after 72 hours (T72). The effect of vehicle alone on cell viability was determined by measuring ATP levels at T0 and 72 hours after treatment of cells with vehicle alone (T72).
  • GI was calculated by applying the following test and equation: If T < V 0 : 100 * 1 - T - V 0 / V 0
    Figure imgb0013
    If T V 0 : 100 * 1 - T - V 0 / V - V 0
    Figure imgb0014
  • Where T is the signal measure for a test article, V is the vehicle-treated control measure, and V0 is the vehicle control measure at time zero. This formula is derived from the Growth Inhibition calculation used in the National Cancer Institute's NCI-60 high throughput screen, as described on the world wide web at dtp.nci.nih.gov/branches/btb/ivclsp.html. A GI reading of 0% represents no growth inhibition-that is, cells treated with compound had similar levels of ATP as cells treated with vehicle and measured at 72 hours. A GI of 100% represents complete growth inhibition-that is, cells treated with compound had similar levels of ATP as cells treated with vehicle and measured at T0. A GI of 100% where cell numbers have not increased during the treatment period is indicative of a cytostatic effect for compounds reaching a plateau at this effect level. A GI of 200% represents complete death of all cells in the culture well which is indicative of an apoptotic effect.
  • For each cell line, the dose-response curve was measured with each HDM2-p53 antagonist and each MEK inhibitor as single agents and in pair wise combinations. Using the potency values from the single agent dose-response curves, the compounds were assigned to different groups of similar potency. Multiple doses above and below the EC50 of a given HDM2-p53 antagonist or MEK inhibitor were measured.
  • Data Normalization
  • In each well of a plate, ATP levels were measured by fluorescence assay as described above. ATP levels are proportional to the number of remaining live cells. This raw data was then normalized in two steps. First, background subtraction was performed by subtracting a mean intensity of negative control wells in which cells were dosed with dimethyl sulfoxide (DMSO). Second, a dynamic range normalization was performed by dividing the background-subtracted data by the mean intensity of positive control wells in which cells were dosed with Staurosporine. The resulting normalized intensity, now with plate-specific effects removed, was used to calculate the Growth Inhibition measure described above.
  • A dose response matrix was constructed where one axis corresponded to one of the three HDM2-p53 antagonists, and the other axis corresponded to one of the MEK inhibitors. The high doses were approximately the given compound's EC90 and were diluted by a constant factor to generate the lower dosage levels. The lowest dose was 0, i.e., the edges of the matrix correspond to another measure of single-agent behavior of each of the compounds. In addition, compounds were combined with themselves in a self-cross analysis for each cell line as described below.
  • Synergy Scores
  • Synergy in general means that two or more compounds, in a given biological context, have a combined effect that is significantly greater than would be expected from the combinations of their single-agent effects. Synergy can be measured in a variety of ways; the methods for measuring synergy involve stating a null hypothesis about the expected single-agent effects, and computing a summary score of the differences between observations and the null hypothesis. A variety of synergy scores in these three broad families were considered.
  • Loewe Synergy, where the null hypothesis is that the two compounds being evaluated are actually the same compound (a self-cross). The effect of combining the two compounds is a function of the sum of concentrations of the two compounds. Significant synergy means the compounds have different effects which work together to create an effect.
  • Bliss Synergy, where the null hypothesis is that the two compounds affect different targets, independently producing the result phenotype so that their effects add like the probability of independent events. Significant synergy means that the compounds somehow work together to produce a net effect, rather than acting independently.
  • Gaddum Synergy, also known as Highest Single Agent Synergy, where the null hypothesis is that there is competitive binding between the two compounds for the same target, and only the compound with the highest binder affinity causes an effect. Significant synergy means that something other than competitive binding is occurring, in which the compounds produce their combined effect in independent ways.
  • Chalice software (Zalicus) was used to perform the bulk of the synergy calculations.
  • Positive Calling Criteria
  • Criteria were imposed to define how much synergy was significant. This was done in four steps:
    • 1: The HDM2-p53 antagonists were self-crossed (each HDM2-p53 antagonist was tested in combination with itself). Synergy scores for each compound measured with itself were determined in each cell line. The resulting synergy scores were typical of false positives (a given compound cannot exhibit synergy with itself). A parametric distribution was fitted to each such population of self-cross synergy scores. For a nonlinear version of Loewe synergy, a gamma distribution was fitted. For most other synergy scores, a normal distribution was fitted, based on the appearance of the histogram of synergy scores and a Shapirio-Wilk test of normality.
      For a false positive rate alpha, a true cross of two distinct compounds were required to have generated a synergy score in the alpha-th tail of the self-cross distribution. Alpha levels of 0.25% to 1% (corresponding to an equivalent normal distribution tail of 2 sigma or 3 sigma), were chosen, depending on the permissiveness of the synergy score. In this way, pairs of compounds in each cell line whose synergy was statistically significantly different from the false positives measured by self-crosses were identified. The p-value measured how far out in the tail of the false positive distribution the potential synergy was.
    • 2: All of the compound pairs were further analyzed using a multiple hypothesis test correction for the group. A Benjamini-Höchberg correction was used, which changed the p-values of step (1) into False Discovery Rates (FDR). A cutoff of 5% or less was imposed on the FDR to obtain an initial set of MEK inhibitors showing synergistic activity in combination with one or more of the HDM2-p53 antagonists.
    • 3: The initial set of MEK inhibitors showing synergistic activity in combination with one or more of the HDM2-p53 antagonists were further filtered by selecting MEK inhibitors that showed synergy with both compound (1) and (2). Compound (3) was not included in the analysis. In order to be declared a positive, it was required that both HDM2-p53 antagonist compounds (1) and (2) had an FDR < 5% with the same MEK inhibitor in the same cell line. Positives were reported by the maximum of the FDR for Compound(1) and Compound (2) (the most stringent call) and the mean of the synergy scores for compounds (1) and (2) (the consensus view).
    • 4: Venn diagram of compound pairs and cell lines that were called as positives was examined for each of the synergy scores used.
  • Thus, a final set of compound pair/cell line positives was chosen based on the nonlinear Loewe synergy, demanding it be in the 5% tail of the self-cross distribution, have a FDR of < 5%, and that Compound (1) and Compound (2) agree on the positive.
  • Enrichment Analysis of Positive Annotation
  • FIG. 1 shows a table of the mean synergy score for Compounds (1) and (2) in combination with the indicated MEK inhibitor in the indicated cell lines. The grey color shows the maximum FDR with Compound (1) and Compound (2) in the indicated cell line. As shown in FIG. 1, MEK inhibitors (6), (7), (8), (9), and (10) showed statistically significant Loewe synergy with both Compound (1) and Compound (2) at an FDR < 5% in some cell lines. The cell lines in which MEK inhibitors synergized with Compounds (1) and (2) appeared to have mutations that activate the RAS pathway signaling such as mutations or Nras, Braf, FLT3-ITD, or EGFR.
  • FIG. 2 shows effect of the combination of Compound (1) and Compound (6) on C32 cells. FIG. 2A shows the mean synergy scores of the combination of Compounds (1) and (6) at the indicated concentrations. FIG. 2B shows percent growth inhibition as a function of concentration of Compound (1), where Compound (6) is not present or is present at 1.1 uM. As shown in FIG. 2, Compound (1) and Compound (6) induce cytostasis of C32 cells when used as single agents. However, the combination of Compound (1) and (6) induced significant cell death.
  • FIG. 3 shows effect of the combination of Compound (1) and Compound (6) on MV4-11 cells. FIG. 2A shows the mean synergy scores of the combination of Compounds (1) and (6) at the indicated concentrations. FIG. 2B shows percent growth inhibition as a function of concentration of Compound (1), where Compound (6) is not present or is present at 3.3 uM. As shown in FIG. 3, Compound (1) induces cytotoxicity in MV4-11 cells and Compound (6) induces cytostasis of MV4-11 cells when used as single agents. However, the combination of Compound (1) and (6) resulted in a cytotoxicity at lower concentrations of Compound (1).
  • FIG. 4 shows effect of the combination of Compound (1) and Compound (6) on A375 cells. FIG. 4A shows the mean synergy scores of the combination of Compounds (1) and (6) at the indicated concentrations. FIG. 4B shows an isobologram of growth inhibition of A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (6), compared to additive growth inhibition. As shown in FIG. 4. the combination of Compounds (1) and (6) displayed synergy in inhibition of A375 cell proliferation. The Loewe Synergy Score for the treatment of A375 cells with the combination of Compounds (1) and (5) is 2.7.
  • FIG. 5 shows effect of the combination of Compound (1) and Compound (5) on Vemurafenib resistant A375 cells. FIG. 5A shows the mean synergy scores of the combination of Compounds (1) and (5) at the indicated concentrations. FIG. 5B shows an isobologram of growth inhibition of Vemurafenib resistant A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (5), compared to additive growth inhibition. As shown in FIG. 5. the combination of Compounds (1) and (5) displayed synergy in inhibition of Vemurafenib resistant A375 cell proliferation. The Loewe Synergy Score for the treatment of Vemurafenib resistant A375 cells with the combination of Compounds (1) and (5) is 2.7.
  • Example 2: Ray Design Analysis Experimental conditions for ATP viability assays:
  • Cells were seeded in 384 well microplates in 47.5 µL of culture medium as described in Table 4 and incubated for 4 hours at 37° C with 5 % CO2. After incubation, 2.5 µL of a 20x concentrated solution of individual test compounds (for single agent tests) or mixtures of test compounds (for combination tests) were added (see "Cell Treatment" section). The microplates are returned to the incubator for 96 h. After incubation, 30 µL of a mix of lysis reagent and luciferase/luciferin was added (CellTiterGlo, Promega) in each well, the microplates were incubated for 1 h at room temperature and the luminescence emitted was measured with a Microbeta Trilux counter (PerkinElmer). Table 4
    Cell line name Culture medium Cell seeding density
    A-375 DMEM, 10% FBS 500
    C32 MEM alpha, 10% FBS 6000
    C8161 DMEM, 10% FBS 600
    DV-90 RPMI 1640, 10% FBS 1000
    HCT116 DMEM, 10% FBS 800
    HT-1197 MEM alpha, 10% FBS 1000
    KU-19-19 RPMI 1640, 10% FBS 600
    NCI-H460 DMEM, 10% FBS 300
    SKMEL-103 RPMI 1640, 10% FBS 1500
    SK-MEL-147 RPMI 1640, 10% FBS 1500
    UACC-62 RPMI 1640, 10% FBS 1000
    UM-UC-3 MEM alpha, 10% FBS 600
  • Cell Treatment:
  • A combination dose response matrix was generated with Compound (1) and Compound (4) or Compound (5) in a 384 well plate. Compound (1) was diluted to generate 14 concentrations and Compound (4) (or Compound (5)) was diluted to generate 18 concentrations. All combinations of concentrations were made in a 14x18 matrix (252 wells). Two identical plates were prepared, allowing duplicate determination of the effect of each combination.
  • In addition, the activity of each of Compound (1), Compound (4) or Compound (5) as a single agent was determined in quadruplicate at the same concentrations, respectively, used in the combination matrix. Calculation of percent inhibition I%: Percent inhibition was calculated for each well according to the formula I % = 100 * T - X / T - B
    Figure imgb0015
  • Where X = the measured value of the well,
  • T = the average value of the control wells containing cells treated with vehicle (DMSO 0.1 %) and B = the average value of blank wells without cells.
  • The 1% values were then used in nonlinear regression analysis to calculate the IC50 of the rays as described in the "Statistical Methodology" section.
  • Experimental design
  • ray design was used to validate findings of synergy. Compound (1) and Compound (4) or Compound (5) were evaluated at several fixed proportions of the compounds in combination. (R. Straetemans, Biometrical journal 47 -2005). The ray design included one ray for each single agent and 19 combination rays. The ray with Compound (1) alone had 14 concentrations of Compound (1), the ray with Compound (4) or Compound (5) alone had 18 concentrations of Compound (4) or Compound (5), and the combination rays had between 7 and 14 concentrations of each compound.
  • Between two and four experiments were performed with each cell line in order to take into account any experimental variability. For each experiment, two 384-well plates were used to obtain duplicates of combination rays and quadruplicates for single agent rays.
  • Statistical methodology
  • Absolute ICX is defined as the concentration of compound where 1% is equal to X%. This measurement allows determining the potential synergistic combinations using the statistical method described hereunder.
  • The relative potency p is first estimated as ρ = ICX A ICX B
    Figure imgb0016
    where ICXA is the ICX of the compound A (here Compound 1) and ICXB is the ICX of the compound B (here Compound 4 or Compound 5). This effective fraction for the ray i is then calculated as f i = 1 c i . ρ + 1
    Figure imgb0017
    where c i = B A
    Figure imgb0018
    is the constant ratio of the concentrations of the compounds A and B in the mixture.
  • Among the combination rays, all the rays for which the effective fraction is in the range 0.05-0.95 are analyzed.
  • A global non linear model using the NLMIXED procedure of the software SAS V9.2 was applied to fit simultaneously the concentration-responses curves for each ray. The model used is a 4-parameter logistic model corresponding to the following equation: Y ikj = Emin i + Emax i - Emin i 1 + exp - m i log Conc ij IC 50 i + ε ijk
    Figure imgb0019
  • Where Yijk is the percentage of inhibition for the kth replicate of the jth concentration in the ith ray
    Concij is the jth mixture concentration (sum of the concentrations of compound A and compound B) in the ith ray
    Emini is the minimum effect obtained from ith ray
    Emaxi is the maximum effect obtained from ith ray
    IC50i is the IC50 obtained from i th ray
    mi is the slope of the curve adjusted with data from ith ray
    εijk is the residual for the kth replicate of the jth concentration in the ith ray, εijk ∼ N(0, σ2)
  • Emin, Emax and/or slope were shared whenever it was possible without degrading the quality of the fit. The combination index Ki of each ray and its 95% confidence interval were then estimated using the following equation based on the Loewe additivity model: C A ICX A + C B ICX B = K i
    Figure imgb0020

    where ICXA and ICXB are the concentrations of compound A and compound B necessary to obtain X% of inhibition for each compound alone and CA and CB are the concentrations of compound A and compound B in the mixture necessary to obtain X% of inhibition. For the combinations studied here, IC50 (or IC40 for compounds with a low activity) have been used to calculate Ki.
  • Additivity for a given Ray was defined as a 95% confidence interval of the Ki that included 1. Significant synergy for a given Ray was defined as a 95% confidence interval of the Ki having an upper bound of less than 1. Significant antagonism was defined as a 95% confidence interval of the Ki having a lower bound of higher than 1.
  • The isobolograms shown in FIGs. 6-17 allow visualization of the position of each Ray. The line shown in FIGs. 6-17 connects the point (0,1) to the point (1,0). All rays below the line correspond to potential synergy, whereas rays above the line correspond to potential antagonism.
  • In the isobolograms shown in FIGs 6-9, and 11-17, the x-axis is [Compound (1)]/IC50[Compound (1)] and the y-axis is [Compound (4)]/IC50[Compound(4)] (FIG. A) or [Compound (5)]/IC50[Compound(5)] (FIG. B). In the isobolograms shown in FIG. 10 the x-axis is [Compound (1)]/IC40[Compound (1)] and the y-axis is [Compound (4)]/IC40[Compound(4)] (FIG. 10A) or [Compound (5)]/IC40[Compound(5)] (FIG. 10B).
  • Synergy was confirmed where the upper bound of the confidence interval of the combination index (Ki) was lower than 1 for the majority of rays. Trend to synergy was confirmed where the combination index (Ki) was lower than 1 (but the confidence interval of the combination index (Ki) included 1) for the majority of rays. Additivity was confirmed where the confidence interval of the combination index (Ki) included 1 for the majority of rays. Trend to antagonism was found where the combination index (Ki) was higher than 1 (but the confidence interval of the combination index (Ki) included 1) for the majority of rays. Antagonism was found where the lower bound of the confidence interval of the combination index (Ki) was higher than 1 for the majority of rays.
  • Compound (1) shows a trend toward antagonism with Compound (4) and additivity with Compound (5) in an Nras mutant melanoma cell line (C8161).
  • Table 5 shows the IC50 estimations for each compound and the relative potency of Compounds (1) and (4) on C8161 cells when used as single agents. Table 5
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) N1 (IC50) 7.74E+01 [6.16E+01 ; 9.72E+01] 1.89E+02 [1.38E+02 ; 2.60E+02] 0.41
    N2 (IC50) N2 (IC50) 1.63E+02 [9.31E+01;2.84E+02] 2.87E+02 [2.24E+02 ; 3.68E+02] 0.57
  • Table 6 shows the interaction characterization of Compounds (1) and (4) in C8161 cells. Table 6
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray
    5 0.05 1.2284 [0.8518 ; 1.7716] Additivity
    N1 Ray
    5 0.07 1.3302 [0.7936 ; 2.2295] Additivity
    N2 Ray
    6 0.15 1.4596 [0.9350 ; 2.2784] Additivity
    N1 Ray
    6 0.20 1.4464 [0.8138 ; 2.5707] Additivity
    N2 Ray
    7 0.35 1.4928 [1.0483 ; 2.1259] Antagonism
    N1 Ray
    7 0.42 1.3625 [0.9226 ; 2.0121] Additivity
    N2 Ray
    8 0.64 1.1827 [0.6624 ; 2.1119] Additivity
    N1 Ray
    8 0.71 1.2192 [0.8524 ; 1.7439] Additivity
    N2 Ray
    9 0.85 1.2099 [0.3842 ; 3.8105] Additivity
    N1 Ray
    9 0.89 1.2529 [0.8711 ; 1.8019] Additivity
    N2 Ray
    10 0.95 0.7767 [0.4027 ; 1.4984] Additivity
    N1 Ray
    10 0.96 1.2059 [0.8505 ; 1.7098] Additivity
  • Table 7 shows the IC50 estimations for each compound and the relative potency of Compounds (1) and (5) on C8161 cells when used as single agents. Table 7
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 1.42E+02 [1.15E+02 ; 1.75E+02] 2.80E+01 [2.13E+01 ; 3.68E+01] 5.07
    N2 (IC50) 9.19E+01 [6.52E+01 ; 1.29E+02] 3.00E+01 [1.83E+01 ; 4.92E+01] 3.06
  • Table 8 shows the interaction characterization of Compounds (1) and (4) in C8161 cells. Table 8
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    7 0.06 0.7853 [0.4884 ; 1.2627] Additivity
    N3 Ray
    6 0.09 1.0619 [0.4659 ; 2.4204] Additivity
    N1 Ray
    8 0.16 0.7728 [0.4591 ; 1.3008] Additivity
    N3 Ray
    7 0.25 1.3380 [0.6550 ; 2.7333] Additivity
    N1 Ray
    9 0.40 1.0063 [0.6789 ; 1.4917] Additivity
    N3 Ray
    8 0.52 1.0316 [0.5872 ; 1.8122] Additivity
    N1 Ray
    10 0.66 0.9705 [0.6897 ; 1.3655] Additivity
    N3 Ray
    9 0.77 1.0863 [0.6502 ; 1.8150] Additivity
    N1 Ray
    11 0.87 0.7584 [0.5434 ; 1.0584] Additivity
    N3 Ray
    10 0.92 0.7807 [0.4402 ; 1.3846] Additivity
    N1 Ray
    12 0.95 1.1543 [0.8403 ; 1.5857] Additivity
    N3 Ray
    11 0.97 0.6960 [0.3887 ; 1.2465] Additivity
  • FIG. 6 shows an isobologram of growth inhibition of C8161 cells treated with Compound (1) in combination with Compound (4) (FIG. 6A) or with Compound (5) (FIG. 6B), compared to additive growth inhibition. As shown in Tables 6 and 8, and FIG. 6, despite the Nras mutant status of C8161 melanoma cells, Compound (1) in combination with Compound (4) showed a trend toward antagonsism and Compound (1) in combination with (5) showed an additive effect on C8161 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in HCT116 Cells
  • Table 9 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on HCT116 cells. Table 9
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 2.96E+02 [1.41E+02 ; 3.63E+02] 1.50E+02 [1.20E+02 ; 1.86E+02] 1.98
    N2 (IC50) 2.96E+02 [2.33E+02 ; 3.77E+02] 1.59E+02 [1.21E+02 ; 2.09E+02] 1.86
    N3 (IC50) 5.30E+02 [3.56E+02 ; 7.89E+02] 4.21E+01 [2.91E+01 ; 6.10E+02] 12.58
    N4 (IC50) 4.92E+02 [3.07E+02 ; 7.87E+02] 6.76E+01 [4.18E+01 ; 1.10E+02] 7.28
  • Table 10 shows the interaction characterization of Compounds (1) and (4) in HCT116 cells. Table 10
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N4 Ray 7 0.04 1.0245 [0.4100 ; 2.5596] Additivity
    N1 Ray 6 0.05 0.5864 [0.3866 ; 0.8896] Synergy
    N2 Ray 6 0.05 0.7371 [0.3785 ; 1.4354] Additivity
    N3 Ray 8 0.07 1.6414 [0.8931 ; 3.0168] Additivity
    N4 Ray 8 0.12 0.9923 [0.4493 ; 2.1919] Additivity
    N1 Ray 7 0.13 0.5293 [0.3563 ; 0.7864] Synergy
    N2 Ray 7 0.14 1.7545 [1.1893 ; 2.5883] Antagonism
    N3 Ray 9 0.21 1.0670 [0.6146 ; 1.8524] Additivity
    N4 Ray 9 0.31 0.6700 [0.3251 ; 1.3808] Additivity
    N1 Ray 8 0.34 0.5892 [0.4156 ; 0.8354] Synergy
    N2 Ray 8 0.35 0.9372 [0.6154 ; 1.4272] Additivity
    N3 Ray 10 0.44 0.6197 [0.3725 ; 1.0310] Additivity
    N4 Ray 10 0.58 0.4061 [0.1945 ; 0.8481] Synergy
    N1 Ray 9 0.63 0.6492 [0.4759 ; 0.8856] Synergy
    N2 Ray 9 0.64 0.7973 [0.5390 ; 1.1795] Additivity
    N3 Ray 11 0.73 0.4591 [0.2767 ; 0.7616] Synergy
    N4 Ray 11 0.82 0.2574 [0.1085 ; 0.6103] Synergy
    N1 Ray 10 0.84 0.7309 [0.5345 ; 0.9995] Synergy
    N2 Ray 10 0.84 0.7431 [0.4950 ; 1.1155] Additivity
    N3 Ray 12 0.89 0.5369 [0.321 ; 0.8958] Synergy
    N4 Ray 12 0.93 0.2159 [0.0810 ; 0.5753] Synergy
    N1 Ray 11 0.94 0.5160 [0.3649 ; 0.7298] Synergy
    N2 Ray 11 0.95 0.5015 [0.3092 ; 0.8133] Synergy
    N3 Ray 13 0.96 0.6990 [0.4083 ; 1.1968] Additivity
  • Table 11 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on HCT116 cells. Table 11
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) N1 (IC50) 3.66E+02 [2.95E+02 ; 4.56E+02] 1.37E+01 [1.08E+01 ; 1.73E+01] 26.84
    N2 (IC50) 2.43E+02 [2.01E+03 ; 2.95E+02] 1.04E+01 [8.38E+00 ; 1.28E+01] 23.49
    N3 (IC50) 6.42E+02 [4.90E+02 ; 8.42E+02] 1.96E+01 [1.49E+01 ; 2.59E+01] 32.75
  • Table 12 shows the interaction characterization of Compounds (1) and (5) in HCT116 cells. Table 12
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N3 Ray
    8 0.03 0.4099 [0.2339 ; 0.7186] Synergy
    N1 Ray
    8 0.04 0.7768 [0.5208 ; 1.1586] Additivity
    N2 Ray
    8 0.04 0.7355 [0.5100 ; 1.0608] Additivity
    N3 Ray
    9 0.09 0.4684 [0.2814 ; 0.7798] Synergy
    N1 Ray
    9 0.11 0.8389 [0.5625 ; 1.2512] Additivity
    N2 Ray
    9 0.12 0.8544 [0.6081 ; 1.2005] Additivity
    N3 Ray
    10 0.23 0.4153 [0.2621 ; 0.6580] Synergy
    N1 Ray
    10 0.27 0.6508 [0.4369 ; 0.9696] Synergy
    N2 Ray
    10 0.30 0.6879 [0.4947 ; 0.9566] Synergy
    N3 Ray
    11 0.50 0.5036 [0.3375 ; 0.7514] Synergy
    N1 Ray
    11 0.55 0.4888 [0.3421 ; 0.6983] Synergy
    N2 Ray
    11 0.59 0.6420 [0.4635 ; 0.8892] Synergy
    N3 Ray
    12 0.75 0.4661 [0.3036 ; 0.7156] Synergy
    N1 Ray
    12 0.79 0.7794 [0.5613 ; 1.0821] Additivity
    N2 Ray
    12 0.81 0.9862 [0.7260 ; 1.3396] Additivity
    N3 Ray
    13 0.91 0.7491 [0.4957 ; 1.1321] Additivity
    N1 Ray
    13 0.93 0.8746 [0.6058 ; 1.2625] Additivity
    N2 Ray
    13 0.93 1.1598 [0.8454 ; 1.5912] Additivity
  • FIG. 7 shows an isobologram of growth inhibition of HCT116 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 7A) or Compound (5) (FIG. 7B), compared to additive growth inhibition. As shown in Tables 10 and 12, and FIG. 7, Compound (1) in combination with Compound (4) or (5) showed a synergistic effect on HCT116 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in an Nras mutant melanoma cell line, SK-MEL-103.
  • Table 13 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on SK-MEL-103 cells. Table 13
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 2.60E+02 [2.11E+02 ; 3.20E+02] 2.85E+01 [2.30E+01 ; 3.53E+01] 9.1
    N2 (IC50) 3.41E+02 [2.62E+02 ; 4.43E+02] 3.16E+01 [2.38E+01 ; 4.18E+01] 10.8
    N3 (IC50) 2.44E+02 [2.10E+02 ; 2.83E+02] 3.40E+01 [2.77E+01 ; 4.17E+01] 7.2
  • Table 14 shows the interaction characterization of Compounds (1) and (4) in SK-MEL-103 cells. Table 14
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    7 0.03 0.5733 [0.3874 ; 0.8483] Synergy
    N3 Ray
    7 0.04 0.7592 [0.5434 ; 1.0606] Additivity
    N2 Ray
    8 0.08 0.9332 [0.5787 ; 1.5047] Additivity
    N1 Ray
    8 0.10 0.7038 [0.4942 ; 1.0022] Additivity
    N3 Ray
    8 0.12 0.8963 [0.6406 ; 1.2542] Additivity
    N2 Ray
    9 0.24 0.7623 [0.4920 ; 1.1811] Additivity
    N1 Ray
    9 0.27 0.6873 [0.4967 ; 0.9511] Synergy
    N3 Ray
    9 0.32 0.5607 [0.4173 ; 0.7534] Synergy
    N2 Ray
    10 0.48 0.6409 [0.4261 ; 0.9640] Synergy
    N1 Ray
    10 0.52 0.6075 [0.4364 ; 0.8457] Synergy
    N3 Ray
    10 0.58 0.6164 [0.4760 ; 0.7983] Synergy
    N2 Ray
    11 0.76 0.5139 [0.3397 ; 0.7776] Synergy
    N1 Ray
    11 0.79 0.6928 [0.5006 ; 0.9588] Synergy
    N3 Ray
    11 0.82 0.6929 [0.5351 ; 0.8971] Synergy
    N2 Ray
    12 0.90 0.5738 [0.3787 ; 0.8694] Synergy
    N1 Ray
    12 0.92 0.7326 [0.5095 ; 1.0536] Additivity
    N3 Ray
    12 0.93 0.7716 [0.6085 ; 0.9785] Synergy
    N2 Ray
    13 0.97 0.6644 [0.4281 ; 1.0311] Additivity
  • Table 15 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on SK-MEL-103 cells. Table 15
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 3.60E+02 [2.82E+02 ; 4.60E+02] 4.83E+00 [3.79E+00 ; 6.16E+00] 74.6
    N2 (IC50) 3.63E+02 [3.00E+02 ; 4.39E+02] 4.58E+00 [3.79E+00 ; 5.54E+00] 79.1
    N3 (IC50) 3.59E+02 [2.82E+02 ; 4.58E+02] 4.47E+00 [3.50E+00 ; 5.73E+00] 80.3
  • Table 16 shows the interaction characterization of Compounds (1) and (5) in SK-MEL-103 cells. Table 16
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N3 Ray
    9 0.04 0.7550 [0.4853 ; 1.1747] Additivity
    N2 Ray
    9 0.04 0.6809 [0.4798 ; 0.9664] Synergy
    N1 Ray
    9 0.04 0.9716 [0.5780 ; 1.6332] Additivity
    N3 Ray
    10 0.11 0.7894 [0.527 ; 1.1816] Additivity
    N2 Ray
    10 0.11 0.6393 [0.4555 ; 0.8974] Synergy
    N1 Ray
    10 0.12 0.5086 [0.3376 ; 0.7662] Synergy
    N3 Ray
    11 0.29 0.5741 [0.4014 ; 0.8209] Synergy
    N2 Ray
    11 0.30 0.5522 [0.4075 ; 0.7485] Synergy
    N1 Ray
    11 0.31 0.5301 [0.3522 ; 0.7979] Synergy
    N3 Ray
    12 0.55 0.6952 [0.5030 ; 0.9610] Synergy
    N2 Ray
    12 0.56 0.6971 [0.5211 ; 0.9326] Synergy
    N1 Ray
    12 0.57 0.6277 [0.4384 ; 0.8987] Synergy
    N3 Ray
    13 0.81 0.8010 [0.565 ; 1.1340] Additivity
    N2 Ray
    13 0.81 0.6753 [0.4990 ; 0.9139] Synergy
    N1 Ray
    13 0.82 0.7837 [0.5361 ; 1.1457] Additivity
    N3 Ray
    14 0.93 0.9801 [0.6760 ; 1.4209] Additivity
    N2 Ray
    14 0.93 0.7425 [0.5427 ; 1.0159] Additivity
    N1 Ray
    14 0.93 0.7598 [0.4911 ; 1.1755] Additivity
    FIG. 8 shows an isobologram of growth inhibition of SK-MEL-103 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 8A) or with Compound (5) (FIG. 8B), compared to additive growth inhibition. As shown in Tables 14 and 16, and FIG. 8, the Compound (1) in combination with Compound (4) or Compound (5) showed a synergistic effect on SK-MEL-103 cells.
  • Compound (1) shows a trend to synergy with Compounds (4) and (5) in UACC-62 cells.
  • Table 17 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on UACC-62 cells. Table 17
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 2.05E+02 [1.51E+02 ; 2.78E+02] 9.87E+00 [7.75E+00; 1.26E+01] 20.8
    N2 (IC50) 3.44E+02 [2.66E+02 ; 4.45E+02] 5.16E+00 [4.17E+00 ; 6.39E+00] 66.72
    N3 (IC50) 1.07E+02 [4.05E+01;2.84E+02] 9.29E+00 [6.57E+00 ; 1.31E+01] 11.55
  • Table 18 shows the interaction characterization of Compounds (1) and (4) in UACC-62 cells. Table 18
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    8 0.05 0.6078 [0.3961 ; 0.9326] synergy
    N2 Ray
    9 0.05 1.2738 [0.8904 ; 1.8222] additivity
    N3 Ray
    8 0.08 1.0897 [0.5725 ; 2.0740] additivity
    N2 Ray
    10 0.13 1.0951 [0.7723 ; 1.5529] additivity
    N1 Ray
    9 0.14 0.5185 [0.3286 ; 0.8182] synergy
    N3 Ray
    9 0.22 0.8687 [0.4733 ; 1.5945] additivity
    N1 Ray
    10 0.32 0.6805 [0.4350 ; 1.0647] additivity
    N2 Ray
    11 0.33 0.7797 [0.5547 ; 1.0960] additivity
    N3 Ray
    10 0.46 0.4797 [0.2268 ; 1.0147] additivity
    N2 Ray
    12 0.60 0.6365 [0.4473 ; 0.9056] synergy
    N1 Ray
    11 0.62 0.5638 [0.3493 ; 0.9098] synergy
    N3 Ray
    11 0.74 0.3222 [0.1202 ; 0.8642] synergy
    N1 Ray
    12 0.83 0.9596 [0.6085 ; 1.5132] additivity
    N2 Ray
    13 0.83 0.6258 [0.4250 ; 0.9216] synergy
    N3 Ray
    12 0.90 0.4484 [0.1384 ; 1.4521] additivity
    N1 Ray
    13 0.94 0.9395 [0.5671 ; 1.5562] additivity
    N2 Ray
    14 0.94 0.5851 [0.3870 ; 0.8846] synergy
    N3 Ray
    13 0.97 0.7522 [0.2092 ; 2.7049] additivity
  • Table 19 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on UACC-62 cells. Table 19
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 1.95E+02 [1.54E+02 ; 2.47E+02] 8.35E-01 [6.76E-01 ; 1.03E+00] 233.36
    N2 (IC50) 1. 12E+02 [7.23E+01 ; 1.72E+02] 1.14E+00 [8.02E-01 ; 1.63E+00] 98.25
    N3 (IC50) 1.32E+02 [9.87E+01 ; 1.77E+02] 1.03E+00 [8.06E-01 ; 1.31E+00] 128.15
  • Table 20 shows the interaction characterization of Compounds (1) and (5) in UACC-62 cells. Table 20
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray
    9 0.03 0.4761 [0.2605 ; 0.8702] Synergy
    N1 Ray
    10 0.04 0.8046 [0.5605 ; 1.1551] Additivity
    N3 Ray
    10 0.07 0.4425 [0.2921 ; 0.6704] synergy
    N2 Ray
    10 0.09 0.4787 [0.2705 ; 0.8470] Synergy
    N1 Ray
    11 0.12 0.6280 [0.4411 ; 0.8942] Synergy
    N3 Ray
    11 0.21 0.6226 [0.4178 ; 0.9278] synergy
    N2 Ray
    11 0.25 0.4913 [0.2781 ; 0.8680] Synergy
    N1 Ray
    12 0.30 0.8162 [0.5788 ; 1.1510] Additivity
    N3 Ray
    12 0.44 0.7460 [0.4982 ; 1.1173] additivity
    N2 Ray
    12 0.51 0.6003 [0.3398 ; 1.0604] Additivity
    N1 Ray
    13 0.59 0.7101 [0.5015 ; 1.0054] Additivity
    N3 Ray
    13 0.72 0.8353 [0.5321 ; 1.3110] additivity
    N2 Ray
    13 0.77 0.8851 [0.4676 ; 1.6753] Additivity
    N1 Ray
    14 0.81 0.5655 [0.3815 ; 0.8381] Synergy
    N3 Ray
    14 0.89 1.0538 [0.6566 ; 1.6912] additivity
    N2 Ray
    14 0.91 0.7710 [0.3855 ; 1.5421] Additivity
    N1 Ray
    15 0.93 0.5147 [0.3444 ; 0.7694] Synergy
    N3 Ray
    15 0.96 0.9601 [0.6005 ; 1.5351] additivity
  • FIG. 9 shows an isobologram of growth inhibition of UACC-62 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 9A) or Compound (5) (FIG. 9B), compared to additive growth inhibition. As shown in Tables 18 and 20, and FIG. 9, Compound (1) in combination with Compound (4) or Compound (5) showed a trend toward synergy on UACC-62 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in HT-1197 cells.
  • Table 21 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on HT-1197 cells. Table 21
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC40) 3.68E+02 [2.04E+02 ; 6.63E+02] 3.81E+02 [1.98E+02 ; 7.35E+02] 0.97
    N2 (IC40) 4.10E+02 [2.10E+02 ; 8.00E+02] 7.30E+01 [3.79E+01 ; 1.41E+02] 5.61
    N3 (IC40) 6.07E+02 [1.93E+02 ; 1.91E+03] 1.30E+02 [5.25E+01 ; 3.20E+02] 4.68
  • Table 22 shows interaction characteristics of Compounds (1) and (4) in HT-1197 cells. Table 22
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray
    7 0.05 0.3150 [0.1005 ; 0.9873] Synergy
    N3 Ray
    7 0.06 0.7615 [0.1413 ; 4.1039] Additivity
    N1 Ray
    6 0.09 0.0724 [0.0242 ; 0.2164] Synergy
    N2 Ray
    8 0.15 0.4372 [0.1668 ; 1.1462] Additivity
    N3 Ray
    8 0.18 0.4460 [0.0992 ; 2.0060] Additivity
    N1 Ray
    7 0.24 0.0285 [0.0110 ; 0.0734] Synergy
    N2 Ray
    9 0.37 0.3530 [0.1568 ; 0.7949] Synergy
    N3 Ray
    9 0.42 0.2049 [0.0444 ; 0.9469] Synergy
    N1 Ray
    8 0.51 0.0531 [0.0216 ; 0.1306] Synergy
    N2 Ray
    10 0.64 0.2341 [0.1043 ; 0.5256] Synergy
    N3 Ray
    10 0.68 0.0377 [0.0101 ; 0.1414] Synergy
    N1 Ray
    9 0.78 0.0805 [0.0354 ; 0.1829] Synergy
    N2 Ray
    11 0.86 0.1562 [0.0659 ; 0.3703] Synergy
    N3 Ray
    11 0.88 0.0499 [0.0131 ; 0.1907] Synergy
    N1 Ray
    10 0.92 0.0640 [0.0249 ; 0.1648] Synergy
    N2 Ray
    12 0.95 0.2129 [0.0875 ; 0.5177] Synergy
    N2 Ray
    12 0.96 0.1648 [0.0370 ; 0.7334] Synergy
    N1 Ray
    11 0.97 0.1867 [0.0745 ; 0.4678] Synergy
  • Table 23 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on HT-1197 cells. Table 23
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC40) 8.91E+02 [3 59E+02 ; 2.21E+03] 8.31E+01 [4.05E+01 ; 1.70E+02] 10.7
    N2 (IC40) 6.13E+02 [2.56E+02 ; 1.47E+03] 1.03E+00 [4.20E+00 ; 2.50E+01] 59.5
    N3 (IC40) 7.30E+02 [4.53E+02 ; 1.18E+03] 1.61E+01 [9.15E+00 ; 2.82E+01] 45.34
    N4 (IC40) 6.02E+02 [3.74E+02 ; 9.68E+02] 1.40E+01 [7.93E+00 ; 2.48E+01] 43
  • Table 24 shows interaction characteristics of Compounds (1) and (5) in HT-1197 cells. Table 24
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray 9 0.05 2.0559 [0.5657 ; 7.4714] Additivity
    N3 Ray 9 0.07 0.2116 [0.0829 ; 0.5403] synergy
    N4 Ray 9 0.07 0.2840 [0.1189 ; 0.6783] synergy
    N1 Ray 8 0.08 0.4962 [0.1566 ; 1.5728] Additivity
    N2 Ray 10 0.14 0.7021 [0.2170 ; 2.2724] Additivity
    N3 Ray 10 0.18 0.1550 [0.0708 ; 0.3392] synergy
    N4 Ray 10 0.19 0.1819 [0.0832 ; 0.3980] synergy
    N1 Ray 9 0.24 0.4944 [0.1796 ; 1.3613] Additivity
    N2 Ray 11 0.36 0.4624 [0.1758 ; 1.2159] Additivity
    N3 Ray 11 0.42 0.0947 [0.0470 ; 0.1910] synergy
    N4 Ray 11 0.44 0.1379 [0.0678 ; 0.2807] synergy
    N1 Ray 10 0.48 0.3983 [0.1617 ; 0.9813] Synergy
    N2 Ray 12 0.63 0.5800 [0.2000 ; 1.6815] Additivity
    N3 Ray 12 0.69 0.0878 [0.0427 ; 0.1804] synergy
    N4 Ray 12 0.70 0.1580 [0.0787 ; 0.3172] synergy
    N1 Ray 11 0.76 0.2001 [0.0632 ; 0.6334] Synergy
    N2 Ray 13 0.85 0.9292 [0.2921 ; 2.9557] Additivity
    N3 Ray 13 0.88 0.1227 [0.0555 ; 0.2712] synergy
    N4 Ray 13 0.89 0.2154 [0.1088 ; 0.4264] synergy
    N1 Ray 12 0.90 0.2923 [0.0875 ; 0.9765] Synergy
    N2 Ray 14 0.94 1.0114 [0.2919 ; 3.5040] Additivity
    N3 Ray 14 0.96 0.1820 [0.0783 ; 0.4229] synergy
    N4 Ray 14 0.96 0.3619 [0.1823 ; 0.7186] synergy
    N1 Ray 13 0.97 0.2589 [0.0638 ; 1.0500] Additivity
  • FIG. 10 shows an isobologram of growth inhibition of HT-1197 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 10A) or Compound (5) (FIG. 10B), compared to additive growth inhibition. As shown in Table 22 and 24 and FIG. 10, Compound (1) in combination with Compound (4) or Compound (5) showed a synergistic effect on HT-1197 cells.
  • Compound (1) shows synergy with Compound (4) and a trend to synergy with Compound (5) in KU-19-19 cells.
  • Table 25 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on KU-19-19 cells. Table 25
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 2.14E+02 [1,66E202 ; 2.077E+02] 4.34E+01 [3.38E+01;5.56E+01] 4.94
    N2 (IC50) 1.78E+02 [1.30E+02 ; 2.44E+02] 3.30E+01 [2.57E3010E4.023E+Ol] 5.40
    N3 (IC50) 2.57E+02 [2.06E+02 ; 3.19E+02] 2.27E+01 [1.90E+01 ; 2.71E+01] 11.3
    N4 (IC50) 2.63E+02 [1.96E+02 ; 3.51E+02] 2.99E+01 [2.32E+01 ; 3.85E+01] 8.79
  • Table 26 shows the interaction characterization of Compounds (1) and (4) in KU-19-19 cells. Table 26
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray 7 0.06 2.0177 [1.1799 ; 3.4504] Antagonism
    N2 Ray 7 0.05 0.5005 [0.3188 ; 0.7860] Synergy
    N3 Ray 8 0.08 1.1001 [0.828 ; 1.4602] additivity
    N4 Ray 8 0.10 0.8438 [0.5576 ; 1.2771] additivity
    N2 Ray 8 0.16 0.5013 [0.3150 ; 0.7978] Synergy
    N1 Ray 8 0.17 0.6707 [0.428 ; 1.0501] Additivity
    N3 Ray 9 0.23 0.9098 [0.7001 ; 1.1822] additivity
    N4 Ray 9 0.27 0.6537 [0.4459 ; 0.9583] synergy
    N2 Ray 9 0.38 0.6125 [0.3899 ; 0.9622] Synergy
    N1 Ray 9 0.40 0.6864 [0.4628 ; 1.0180] Additivity
    N3 Ray 10 0.47 0.6994 [0.5401 ; 0.9056] synergy
    N4 Ray 10 0.53 0.4986 [0.3424 ; 0.7260] synergy
    N2 Ray 10 0.65 0.3878 [0.2425 ; 0.6201] Synergy
    N1 Ray 10 0.67 0.7675 [0.5348 ; 1.1014] Additivity
    N3 Ray 11 0.75 0.5599 [0.4220 ; 0.7429] synergy
    N4 Ray 11 0.79 0.4680 [0.3084 ; 0.7104] synergy
    N2 Ray 11 0.86 0.2520 [0.1560 ; 0.4070] Synergy
    N1 Ray 11 0.87 0.4106 [0.2679 ; 0.6292] Synergy
    N3 Ray 12 0.90 0.5630 [0.4159 ; 0.7621] synergy
    N4 Ray 12 0.92 0.5323 [0.3356 ; 0.8441] synergy
    N1 Ray 12 0.95 0.7081 [0.4705 ; 1.0656] Additivity
    N2 Ray 12 0.95 0.9759 [0.6214 ; 1.5325] Additivity
    N3 Ray 13 0.97 0.6125 [0.4421 ; 0.8487] synergy
    N4 Ray 13 0.97 0.6048 [0.3705 ; 0.9874] synergy
  • Table 27 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on KU-19-19 cells. Table 27
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 2.80E+02 [2.07E+02 ; 3.79E+02] 2.60E+01 [1.95E+01 ; 3.46E+01] 10.77
    N2 (IC50) 3.38+02 [2.20E+02 ; 5.19E+02] 3.04E+00 [2.07E+00 ; 4.48E+00] 111.18
    N3 (IC50) 2.82E+02 [2.12E+02 ; 3.76E+02] 2.42E+00 [1.98E+00 ; 2.95E+00] 116.62
    N4 (IC50) 1.94E+02 [1.53E+02 ; 2.47E+02] 2.33E+00 [1.94E+00 ; 2.79E+00] 83.34
  • Table 28 shows the interaction characterization of Compounds (1) and (5) in KU-19-19 cells. Table 28
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray 10 0.08 1.7399 [0.7995 ; 3.7864] additivity
    N3 Ray 10 0.08 0.8535 [0.6111 ; 1.1920] additivity
    N1 Ray 8 0.09 1.0924 [0.6788 ; 1.7580] additivity
    N4 Ray 10 0.11 0.8316 [0.6105 ; 1.1327] additivity
    N3 Ray 11 0.22 0.6394 [0.4607 ; 0.8873] synergy
    N2 Ray 11 0.23 1.4696 [0.7745 ; 2.7885] additivity
    N1 Ray 9 0.24 0.8856 [0.5602 ; 1.3999] additivity
    N4 Ray 11 0.29 0.7753 [0.5830 ; 1.0311] additivity
    N3 Ray 12 0.46 0.5302 [0.3765 ; 0.7467] synergy
    N2 Ray 12 0.47 1.5136 [0.8200 ; 2.7941] additivity
    N1 Ray 10 0.48 0.8336 [0.5448 ; 1.2755] additivity
    N4 Ray 12 0.55 0.8118 [0.6135 ; 1.0742] additivity
    N3 Ray 13 0.74 0.4581 [0.307 ; 0.6816] synergy
    N2 Ray 13 0.75 1.0034 [0.5070 ; 1.9860] additivity
    N1 Ray 11 0.76 0.4396 [0.2727 ; 0.7087] synergy
    N4 Ray 13 0.80 0.8254 [0.6023 ; 1.1312] additivity
    N2 Ray 14 0.90 0.9596 [0.4619 ; 1.9938] additivity
    N1 Ray 12 0.90 0.6701 [0.4117 ; 1.0907] additivity
    N3 Ray 14 0.90 0.4489 [0.2910 ; 0.6922] synergy
    N4 Ray 14 0.92 0.8246 [0.5809 ; 1.1705] additivity
    N1 Ray 13 0.97 0.7644 [0.4450 ; 1.3130] additivity
    N2 Ray 15 0.97 0.7806 [0.3535 ; 1.7238] additivity
    N3 Ray 15 0.97 0.4715 [0.3011 ; 0.7383] synergy
    N4 Ray 15 0.98 0.6628 [0.4572 ; 0.9608] synergy
  • FIG. 11 shows an isobologram of growth inhibition of KU-19-19 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 11A) or Compound (5) (FIG. 11B), compared to additive growth inhibition. As shown in Tables 26 and 28, and FIG. 11, Compound (1) in combination with Compound (4) showed a synergistic effect on KU-19-19 cells and Compound (1) in combination with Compound (5) showed a trend to synergy.
  • Compound (1) shows additivity with Compound (4) and Compound (5) in a Braf mutant melanoma cell line, A375.
  • Table 29 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on A375 cells. Table 29
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 3.10E+02 [2.67E+02 ; 3.60E+02] 1.01E+01 [8.64E+00 ; 1.18E+01] 30.7
    N2 (IC50) 2.88E+02 [2.41E+02 ; 3.45E+02] 1.12E+01 [9.32E+00 ; 1.35E+01] 25.7
  • Table 30 shows the interaction characteristics of Compounds (1) and (4) in A375 cells. Table 30
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    8 0.03 1.0838 [0.8505 ; 1.3812] Additivity
    N2 Ray
    8 0.04 0.9608 [0.6980 ; 1.3228] Additivity
    N1 Ray
    9 0.10 0.9830 [0.7610 ; 1.2697] Additivity
    N2 Ray
    9 0.11 1.1334 [0.8346 ; 1.5392] Additivity
    N1 Ray
    10 0.25 1.0991 [0.8656 ; 1.3957] Additivity
    N2 Ray
    10 0.28 1.1329 [0.8649 ; 1.4839] Additivity
    N1 Ray
    11 0.52 1.1522 [0.9368 ; 1.4171] Additivity
    N2 Ray
    11 0.56 1.028 [0.7835 ; 1.3487] Additivity
    N1 Ray
    12 0.76 0.9289 [0.7305 ; 1.1811] Additivity
    N2 Ray
    12 0.80 1.2189 [0.9297 ; 1.5982] Additivity
    N1 Ray
    13 0.92 0.7160 [0.5463 ; 0.9384] Synergy
    N2 Ray
    13 0.93 1.0725 [0.7907 ; 1.4548] Additivity
  • Table 31 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on A375 cells. Table 31
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 2.64E+02 [2.38E+02 ; 2.92E+02] 7.01E-01 [6.32E-01 ; 7.77E-01] 376.3
    N2 (IC50) 2.89E+02 [2.41E+02 ; 3.47E+02] 9.26E-01 [7.81E-01 ; 1.10E+00] 312.3
  • Table 32 shows the interaction characteristics of Compounds (1) and (5) in A375 cells. Table 32
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray
    10 0.03 1.8132 [1.3613 ; 2.4153] Antagonism
    N1 Ray
    11 0.08 1.1294 [0.9616 ; 1.3265] Additivity
    N2 Ray
    11 0.10 0.8676 [0.6326 ; 1.1899] Additivity
    N1 Ray
    12 0.21 1.2616 [1.0814 ; 1.4717] Antagonism
    N2 Ray
    12 0.24 1.1801 [0.9184 ; 1.5164] Additivity
    N1 Ray
    13 0.47 1.1666 [1.0061 ; 1.3528] Antagonism
    N2 Ray
    13 0.52 1.1023 [0.8396 ; 1.4471] Additivity
    N1 Ray
    14 0.73 1.2623 [1.0831 ; 1.4711] Antagonism
    N2 Ray
    14 0.76 1.1830 [0.9087 ; 1.5399] Additivity
    N1 Ray
    15 0.90 1.0731 [0.9103 ; 1.2651] Additivity
    N2 Ray
    15 0.91 0.8740 [0.6238 ; 1.2248] Additivity
    N1 Ray
    16 0.96 1.1028 [0.9311 ; 1.3063] Additivity
  • FIG. 12 shows an isobologram of growth inhibition of A375 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 12A) or with Compound (5) (FIG. 12B) compared to additive growth inhibition. As shown in Tables 30 and 32, and FIG. 12, Compound (1) in combination with Compound (4) or (5) showed an additive effect on A375 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in Braf mutant melanoma cells, C32.
  • Table 33 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on C32 cells. Table 33
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 8.13E+02 [5.86E+02 ; 1.13E+03] 1.74E+01 [1.47E+01 ; 2.06E+01] 46.7
    N2 (IC50) 9.62E+02 [6.75E+02 ; 1.37E+03] 2.36E+01 [2.03E+01 ; 2.74E+01] 40.8
    N3 (IC50) 1.45E+03 [1.19E+03 ; 1.77E+03] 2.27E+01 [2.03E+01 ; 2.54E+01] 64.1
  • Table 34 shows the interaction characteristics of Compounds (1) and (4) in C32 cells. Table 34
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N3 Ray
    9 0.05 0.7614 [0.6225 ; 0.9313] Synergy
    N1 Ray
    9 0.07 0.7740 [0.5815 ; 1.0303] Additivity
    N2 Ray
    9 0.08 0.8382 [0.6282 ; 1.1183] Additivity
    N3 Ray
    10 0.14 0.6935 [0.5733 ; 0.8388] Synergy
    N1 Ray
    10 0.18 0.7019 [0.5210 ; 0.9457] Synergy
    N2 Ray
    10 0.20 0.6164 [0.4593 ; 0.8272] Synergy
    N3 Ray
    11 0.34 0.5301 [0.4268 ; 0.6585] Synergy
    N1 Ray
    11 0.42 0.6887 [0.4903 ; 0.9673] Synergy
    N2 Ray
    11 0.45 0.5207 [0.3702 ; 0.7324] Synergy
    N3 Ray
    12 0.61 0.5798 [0.4516 ; 0.7444] Synergy
    N1 Ray
    12 0.68 0.7610 [0.5068 ; 1.1427] Additivity
    N2 Ray
    12 0.71 0.7183 [0.4802 ; 1.0744] Additivity
    N3 Ray
    13 0.84 0.6554 [0.4819 ; 0.8912] Synergy
    N1 Ray
    13 0.88 0.9736 [0.6169 ; 1.5366] Additivity
    N2 Ray
    13 0.89 0.9132 [0.5504 ; 1.5150] Additivity
    N3 Ray
    14 0.94 0.7955 [0.5759 ; 1.0988] Additivity
    N1 Ray
    14 0.96 1.2713 [0.7803 ; 2.0713] Additivity
    N2 Ray
    14 0.96 1.3182 [0.7815 ; 2.2235] Additivity
  • Table 35 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on C32 cells. Table 35
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC40) 8.23E+02 [4.42E+02 ; 1.53E+03] 1.13E+00 [9.89E-01 ; 1.29E+00] 727
    N2 (IC40) 6.18E+02 [3.44E+02 ; 1.11E+03] 1.19E+00 [9.79E-01 ; 1.45E+00] 518.5
    N3 (IC40) 2.15E+03 [1.77E+03 ; 2.62E+03] 8.46E-01 [7.06E-01 ; 1.02E+00] 2545.0
    N4 (IC40) 1.07E+03 [7.24E+02 ; 1.57E+03] 7.88E-01 [5.93E-01 ; 1.05E+00] 1353.5
  • Table 36 shows the interaction characteristics of Compounds (1) and (5) in C32 cells. Table 36
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N4 Ray
    11 0.02 0.7309 [0.4502 ; 1.1867] Additivity
    N1 Ray
    11 0.04 0.6662 [0.5318 ; 0.8347] Synergy
    N3 Ray
    12 0.04 0.9525 [0.7258 ; 1.2500] Additivity
    N2 Ray
    11 0.06 0.7353 [0.5151 ; 1.0497] Additivity
    N4 Ray
    12 0.07 0.5630 [0.3466 ; 0.9144] Synergy
    N1 Ray
    12 0.12 0.8010 [0.6482 ; 0.9899] Synergy
    N3 Ray
    13 0.12 0.6808 [0.5177 ; 0.8954] Synergy
    N2 Ray
    12 0.16 0.7696 [0.5376 ; 1.1018] Additivity
    N4 Ray
    13 0.20 0.6761 [0.407 ; 1.1206] Additivity
    N3 Ray
    14 0.28 0.5832 [0.4382 ; 0.7762] Synergy
    N1 Ray
    13 0.31 0.6753 [0.5002 ; 0.9118] Synergy
    N2 Ray
    13 0.39 0.6700 [0.4271 ; 1.0509] Additivity
    N4 Ray
    14 0.42 0.4637 [0.2750 ; 0.7819] Synergy
    N3 Ray
    15 0.57 0.6566 [0.4961 ; 0.8690] Synergy
    N2 Ray
    14 0.66 1.0079 [0.5921 ; 1.7157] Additivity
    N4 Ray
    15 0.71 0.5161 [0.3036 ; 0.8772] Synergy
    N3 Ray
    16 0.80 0.8643 [0.6417 ; 1.1639] Additivity
    N1 Ray
    15 0.82 0.9326 [0.5200 ; 1.6726] Additivity
    N2 Ray
    15 0.87 1.5207 [0.8099 ; 2.8552] Additivity
    N1 Ray
    16 0.93 0.6917 [0.3420 ; 1.3991] Additivity
    N3 Ray 17 0.93 1.0378 [0.7652 ; 1.4075] Additivity
  • FIG. 13 shows an isobologram of growth inhibition of C32 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 13A) or with Compound (5) (FIG. 13B) compared to additive growth inhibition. As shown in Tables 34 and 36, and FIG. 13, Compound (1) in combination with Compound (4) or Compound (5) showed a synergistic effect on C32 cells.
  • Compound (1) shows synergy with Compounds (4) and (5) in SK-MEL-147 cells.
  • Table 37 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (4) on SK-MEL-147 cells. Table 37
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 5.56E+02 [3.97E+02 ; 7.77E+02] 9.12E+01 [6.45E+01 ; 1.29E+02] 6.1
    N2 (IC50) 8.78E+02 [6.56E+02; 1.17E+03] 8.24E+01 [5.99E+01 ; 1.13E+02] 10.7
    N3 (IC50) 8.66E+02 [6.57E+02; 1.14E+03] 8.14E+01 [6.08E+01 ; 1.09E+02] 10.6
  • Table 38 shows the interaction characteristics of Compounds (1) and (4) in SK-MEL-147 cells. Table 38
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray
    7 0.03 0.8443 [0.4859 ; 1.4671] Additivity
    N1 Ray
    7 0.05 0.6337 [0.3663 ; 1.0965] Additivity
    N2 Ray
    8 0.09 0.6180 [0.3662 ; 1.0430] Additivity
    N3 Ray
    8 0.09 0.6558 [0.3798 ; 1.1323] Additivity
    N1 Ray
    8 0.14 0.3892 [0.228 ; 0.6636] Synergy
    N2 Ray
    9 0.24 0.6069 [0.3901 ; 0.9443] Synergy
    N3 Ray
    9 0.24 0.2878 [0.1692 ; 0.4896] Synergy
    N1 Ray
    9 0.35 0.4817 [0.2814 ; 0.8248] Synergy
    N2 Ray
    10 0.48 0.7068 [0.4690 ; 1.0652] Additivity
    N3 Ray
    10 0.48 0.4086 [0.2603 ; 0.6413] Synergy
    N1 Ray
    10 0.62 0.4286 [0.2576 ; 0.7131] Synergy
    N2 Ray
    11 0.76 0.5105 [0.3279 ; 0.7947] Synergy
    N3 Ray
    11 0.76 0.4263 [0.2726 ; 0.6668] Synergy
    N1 Ray
    11 0.85 0.4476 [0.2623 ; 0.7640] Synergy
    N2 Ray
    12 0.90 0.6678 [0.4270 ; 1.0444] Additivity
    N3 Ray
    12 0.90 0.5595 [0.3526 ; 0.8877] Synergy
    N1 Ray
    12 0.94 0.5124 [0.2948 ; 0.8908] Synergy
    N3 Ray
    13 0.97 0.6469 [0.3981 ; 1.0512] Additivity
  • Table 39 shows the IC50 estimations for each compound alone and the relative potency of Compounds (1) and (5) on SK-MEL-147 cells. Table 39
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 9.78E+02 [7.39E+02 ; 1.29E+03] 6.94E+00 [5.39E+00 ; 8.93E+00] 141.0
    N2 (IC50) 9.68E+02 [7.93E+02 ; 1.18E+03] 6.94E+00 [5.62E+00 ; 8.57E+] 139.4
    N3 (IC50) 1.08E+03 [8.80E+02 ; 1.32E+03] 7.42E+00 [6.05E+00 ; 9.10E+00] 145.2
  • Table 40 shows the interaction characteristics of Compounds (1) and (5) in SK-MEL-147 cells. Table 40
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N3 Ray
    10 0.06 0.6569 [0.4573 ; 0.9436] Synergy
    N1 Ray
    10 0.07 0.6580 [0.4227 ; 1.0243] Additivity
    N2 Ray
    10 0.07 0.8604 [0.6040 ; 1.2258] Additivity
    N1 Ray
    11 0.19 0.5333 [0.3346 ; 0.8498] Synergy
    N2 Ray
    11 0.19 0.5885 [0.4167 ; 0.8311] Synergy
    N3 Ray
    11 0.19 0.5846 [0.4240 ; 0.8061] Synergy
    N1 Ray
    12 0.41 0.6460 [0.4447 ; 0.9383] Synergy
    N3 Ray
    12 0.41 0.4436 [0.3235 ; 0.6084] Synergy
    N2 Ray
    12 0.42 0.5921 [0.4299 ; 0.8156] Synergy
    N1 Ray
    13 0.70 0.5642 [0.3659 ; 0.8702] Synergy
    N3 Ray
    13 0.70 0.4736 [0.3484 ; 0.6439] Synergy
    N2 Ray
    13 0.71 0.4888 [0.3511 ; 0.6804] Synergy
    N3 Ray
    14 0.87 0.5728 [0.4078 ; 0.8047] Synergy
    N1 Ray
    14 0.88 0.7568 [0.4829 ; 1.1860] Additivity
    N2 Ray
    14 0.88 0.6583 [0.469 ; 0.9222] Synergy
    N1 Ray
    15 0.96 0.8858 [0.5535 ; 1.4177] Additivity
    N2 Ray
    15 0.96 0.7126 [0.5018 ; 1.0120] Additivity
    N3 Ray
    15 0.96 0.7981 [0.5680 ; 1.1215] Additivity
  • FIG. 14 shows an isobologram of growth inhibition of SK-MEL-147 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 14A) or Compound (5) (FIG. 14B), compared to additive growth inhibition. As shown in Tables 38 and 40, and FIG. 14, the combination of Compound (1) and Compound (4) or (5) showed a synergistic effect on SK-MEL-147 cells.
  • Compound (1) shows additivity with Compound (4) in DV90 cells.
  • Table 41 shows the IC50 estimations for each compound used alone and relative potency of Compounds (1) and (4) on DV90 cells. Table 41
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 1.40E+02 [6.90E+01 ; 2.85E+02] 3.64E+01 [2.10E+01 ; 6.30E+01] 3.9
    N2 (IC50) 1.63E+02 [9.22E+01 ; 2.87E+02] 1.97E+01 [9.77E+00 ; 3.99E+01] 8.2
    N3 (IC50) 2.54E+02 [1.48E+02 ; 4.35E+02] 5.55E+01 [3.23E+01 ; 9.52E+01] 4.6
  • Table 42 shows the interaction characteristics of Compounds (1) and (4) in DV90 cells. Table 42
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    6 0.03 1.1814 [0.428 ; 3.2561] Additivity
    N2 Ray
    7 0.04 1.8696 [0.5511 ; 6.3424] Additivity
    N3 Ray
    7 0.06 0.4777 [0.1699 ; 1.3435] Additivity
    N1 Ray
    7 0.07 0.7219 [0.2082 ; 2.5032] Additivity
    N2 Ray
    8 0.11 0.9143 [0.31999; 2.6131] Additivity
    N3 Ray
    8 0.18 1.4945 [0.5946 ; 3.7567] Additivity
    N1 Ray
    8 0.21 0.7855 [0.2519 ; 2.4499] Additivity
    N2 Ray
    9 0.29 1.0474 [0.3818 ; 2.8733] Additivity
    N3 Ray
    9 0.42 0.6976 [0.2907 ; 1.6745] Additivity
    N1 Ray
    9 0.46 0.8797 [0.3443 ; 2.2477] Additivity
    N2 Ray
    10 0.55 0.7820 [0.3678 ; 1.6623] Additivity
    N3 Ray
    10 0.69 0.4475 [0.1569 ; 1.2760] Additivity
    N1 Ray
    10 0.72 0.4393 [0.164 ; 1.1715] Additivity
    N2 Ray
    11 0.80 1.4445 [0.6724 ; 3.1029] Additivity
    N3 Ray
    11 0.88 0.3989 [0.1488 ; 1.0693] Additivity
    N1 Ray
    11 0.90 0.4137 [0.1524 ; 1.1232] Additivity
    N2 Ray
    12 0.92 1.4103 [0.5895 ; 3.3738] Additivity
    N3 Ray
    12 0.96 0.7001 [0.307 ; 1.5920] Additivity
  • Table 43 shows the IC50 estimations for each compound used alone and relative potency of Compounds (1) and (5) on DV90 cells. Table 43
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 1.39E+02 [8.65E+01 ; 2.23E+02] 3.23E+00 [2.20E+00 ; 4.72E+00] 43.1
    N2 (IC50) 7.83E+01 [3.74E+01 ; 1.64E+02] 1.20E+00 [6.47E-01 ; 2.24E+00] 65.0
    N3 (IC50) 2.07E+02 [1.57E+02 ; 2.72E+02] 9.03E-01 [4.62E-01 ; 1.77E+00] 228.9
  • Table 44 shows the interaction characteristics of Compounds (1) and (5) in DV90 cells. Table 44
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N3 Ray
    10 0.04 3.2416 [1.5428 ; 6.8111] Antagonism
    N2 Ray
    9 0.05 0.4163 [0.1210 ; 1.4320] Additivity
    N1 Ray
    9 0.07 1.6379 [0.8013 ; 3.3482] Additivity
    N2 Ray
    10 0.13 0.8221 [0.3180 ; 2.1249] Additivity
    N3 Ray
    11 0.13 2.6946 [1.3277 ; 5.4687] Antagonism
    N1 Ray
    10 0.19 0.8337 [0.4408 ; 1.5766] Additivity
    N3 Ray
    12 0.30 1.6455 [0.9386 ; 2.8849] Additivity
    N2 Ray
    11 0.34 0.1919 [0.0804 ; 0.4583] Synergy
    N1 Ray
    11 0.44 0.6070 [0.3066 ; 1.2017] Additivity
    N3 Ray
    13 0.59 1.0816 [0.6800 ; 1.7205] Additivity
    N2 Ray
    12 0.61 0.0795 [0.0323 ; 0.1954] Synergy
    N1 Ray
    12 0.70 0.5166 [0.2706 ; 0.9862] Synergy
    N3 Ray
    14 0.81 1.2789 [0.8622 ; 1.8971] Additivity
    N2 Ray
    13 0.84 0.0936 [0.0319 ; 0.2745] Synergy
    N1 Ray
    13 0.89 0.8042 [0.4102 ; 1.5764] Additivity
    N2 Ray
    14 0.94 0.1161 [0.0412 ; 0.3278] Synergy
    N3 Ray
    15 0.94 0.9426 [0.5601 ; 1.5863] Additivity
    N1 Ray
    14 0.96 1.3607 [0.6770 ; 2.7348] Additivity
  • FIG. 15 shows an isobologram of growth inhibition of DV90 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 15A) or Compound (5) (FIG. 15B), compared to additive growth inhibition. As shown in Tables 42 and 43, and FIG. 15, Compound (1) in combination with Compound (4) showed additivity on DV90 cells.
  • Compound (1) shows synergy with Compound (5) in H460 cells.
  • Table 45 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on H460 cells. Table 45
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 3.19E+02 [2.61E+02 ; 3.89E+02] 1.55E+02 [1.07E+02 ; 2.24E+02] 2.1
    N2 (IC50) 1.66E+02 [1.25E+02 ; 2.20E+02] 8.40E+01 [6.20E+01 ; 1.14E+02] 2.0
    N3 (IC50) 1.36E+02 [9.00E+01 ; 2.07E+02] 1.04E+02 [6.48E+01 ; 1.68E+02] 1.3
  • Table 46 shows the interaction characteristics of Compounds (1) and (4) in H460 cells. Table 46
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    6 0.05 1.5331 [0.9970 ; 2.3574] Additivity
    N2 Ray
    6 0.05 0.7694 [0.3938 ; 1.5031] Additivity
    N3 Ray
    6 0.07 0.7949 [0.3405 ; 1.8555] Additivity
    N1 Ray
    7 0.13 0.7955 [0.4571 ; 1.3843] Additivity
    N2 Ray
    7 0.13 1.1173 [0.6732 ; 1.8545] Additivity
    N3 Ray
    7 0.19 0.7763 [0.3564 ; 1.6910] Additivity
    N1 Ray
    8 0.33 0.5376 [0.3261 ; 0.8862] Synergy
    N2 Ray
    8 0.34 1.3456 [0.8795 ; 2.0587] Additivity
    N3 Ray
    8 0.43 1.7012 [0.8964 ; 3.2285] Additivity
    N1 Ray
    9 0.62 0.6094 [0.3864 ; 0.9611] Synergy
    N2 Ray
    9 0.63 0.5693 [0.3675 ; 0.8819] Synergy
    N3 Ray
    9 0.72 0.4729 [0.2427 ; 0.9214] Synergy
    N1 Ray
    10 0.83 0.6137 [0.3982 ; 0.9456] Synergy
    N2 Ray
    10 0.83 1.0664 [0.7122 ; 1.5966] Additivity
    N3 Ray
    10 0.88 0.4807 [0.2338 ; 0.9886] Synergy
    N1 Ray
    11 0.94 1.4303 [1.0277 ; 1.9907] Antagonism
    N2 Ray
    11 0.94 1.0616 [0.6981 ; 1.6144] Additivity
    N3 Ray
    11 0.96 0.8813 [0.4655 ; 1.6684] Additivity
  • Table 47 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on H460 cells. Table 47
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 2.53E+02 [2.15E+02 ; 2.99E+02] 6.82E+00 [4.95E+00 ; 9.39E+00] 37.1
    N2 (IC50) 3.19E+02 [2.72E+02 ; 3.75E+02] 1.01E+01 [7.69E+00 ; 1.34E+01] 31.5
    N3 (IC50) 2.97E+02 [2.29E+02 ; 3.87E+02] 1.10E+01 [7.67E+00 ; 1.59E+01] 26.9
  • Table 48 shows the interaction characteristics of Compounds (1) and (5) in H460 cells. Table 48
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N3 Ray
    8 0.04 1.7372 [0.9459 ; 3.1903] Additivity
    N2 Ray
    9 0.10 0.7189 [0.4787 ; 1.0795] Additivity
    N3 Ray
    9 0.11 0.7200 [0.4072 ; 1.2730] Additivity
    N1 Ray
    10 0.21 0.7177 [0.4766 ; 1.0808] Additivity
    N2 Ray
    10 0.24 0.7328 [0.5205 ; 1.0318] Additivity
    N3 Ray
    10 0.27 0.9753 [0.6162 ; 1.5438] Additivity
    N1 Ray
    11 0.47 0.6777 [0.4884 ; 0.9405] Synergy
    N2 Ray
    11 0.51 0.6608 [0.4894 ; 0.8923] Synergy
    N3 Ray
    11 0.55 0.2378 [0.1355 ; 0.4172] Synergy
    N1 Ray
    12 0.73 0.6800 [0.504 ; 0.9166] Synergy
    N2 Ray
    12 0.76 0.6296 [0.4807 ; 0.8246] Synergy
    N3 Ray
    12 0.79 0.4957 [0.3146 ; 0.7811] Synergy
    N1 Ray
    13 0.90 0.7058 [0.5186 ; 0.9607] Synergy
    N2 Ray
    13 0.91 0.6739 [0.4993 ; 0.9095] Synergy
    N3 Ray
    13 0.93 0.8401 [0.5394 ; 1.3085] Additivity
    N1 Ray
    14 0.96 0.9734 [0.7417 ; 1.2775] Additivity
    N2 Ray
    14 0.97 0.7608 [0.5925 ; 0.9770] Synergy
  • FIG. 16 shows an isobologram of growth inhibition of H460 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 16A) or Compound (5) (FIG. 16B), compared to additive growth inhibition. As shown in Tables 46 and 48, and FIG. 16, Compound (1) in combination with Compound (5) showed a synergistic effect on H460 cells.
  • Compound (1) shows additivity with Compounds (4) and (5) in UM-UC-3 cells.
  • Table 49 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (4) on UM-UC-3 cells. Table 49
    Experiment Compound 1 Compound 4 Relative potency
    N1 (IC50) 8.53E+03 [6.54E+03 ; 1.11E+04] 2.63E+02 [1.53E+02 ; 4.52E+02] 32.48
    N2 (IC50) 6.00E+03 [4.86E+03 ; 7.41E+03] 7.37E+01 [4.92E+01 ; 1.10E+02] 81.43
  • Table 50 shows the interaction characteristics of Compounds (1) and (4) in UM-UC-3 cells. Table 50
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N2 Ray
    9 0.04 1.7681 [0.9837; 3.1780] Additivity
    N1 Ray
    9 0.09 1.2061 [0.5751; 2.5293] Additivity
    N2 Ray
    10 0.11 1.0148 [0.5706; 1.8047] Additivity
    N1 Ray
    10 0.24 0.7708 [0.4036; 1.4723] Additivity
    N2 Ray
    11 0.29 0.6674 [0.3921; 1.1360] Additivity
    N1 Ray
    11 0.51 0.6154 [0.3304; 1.1462] Additivity
    N2 Ray
    12 0.55 0.9561 [0.7191; 1.2711] Additivity
    N1 Ray
    12 0.75 0.7632 [0.4881; 1.1933] Additivity
    N2 Ray
    13 0.80 0.8798 [0.6518; 1.1875] Additivity
    N1 Ray
    13 0.91 0.9695 [0.6166; 1.5244] Additivity
    N2 Ray
    14 0.92 0.9271 [0.6730; 1.2770] Additivity
    N1 Ray
    14 0.97 0.9708 [0.6301; 1.4957] Additivity
  • Table 51 shows the IC50 estimations for each compound alone and relative potency of Compounds (1) and (5) on UM-UC-3 cells. Table 51
    Experiment Compound 1 Compound 5 Relative potency
    N1 (IC50) 6.95E+03 [5.65E+03 ; 8.55E+03] 2.24E+02 [1.31E+02 ; 3.85E+02] 31.0
    N2 (IC50) 5.40E+03 [4.54E+03 ; 6.43E+03] 2.34E+01 [1.65E+01 ; 3.33E+01] 230.5
    N3 (IC50) 5.75E+03 [4.52E+03 ; 7.31E+03] 2.30E+01 [1.43E+01 ; 3.70E+01] 249.7
  • Table 52 shows the interaction characteristics of Compounds (1) and (5) in UM-UC-3 cells. Table 52
    Experiment Ray f values Ki (confidence interval at 95%) Interaction characterization
    N1 Ray
    8 0.03 1.0778 [0.4454 ; 2.6084] Additivity
    N2 Ray
    10 0.04 0.9437 [0.5595 ; 1.5917] Additivity
    N3 Ray
    10 0.04 1.1040 [0.5600 ; 2.1765] Additivity
    N1 Ray
    9 0.10 0.9785 [0.4360 ; 2.1962] Additivity
    N3 Ray
    11 0.12 0.8211 [0.4481 ; 1.5044] Additivity
    N2 Ray
    11 0.13 0.8543 [0.5354 ; 1.3630] Additivity
    N1 Ray
    10 0.24 1.4840 [0.6757 ; 3.2592] Additivity
    N3 Ray
    12 0.29 0.6503 [0.3812 ; 1.1094] Additivity
    N2 Ray
    12 0.30 0.8290 [0.5545 ; 1.2395] Additivity
    N1 Ray
    11 0.52 0.8182 [0.4922 ; 1.3602] Additivity
    N3 Ray
    13 0.57 0.7347 [0.4836 ; 1.1160] Additivity
    N2 Ray
    13 0.59 0.8713 [0.6481 ; 1.1715] Additivity
    N1 Ray
    12 0.76 0.7628 [0.5120 ; 1.1364] Additivity
    N3 Ray
    14 0.80 0.7493 [0.5542 ; 1.0131] Additivity
    N2 Ray
    14 0.81 0.9370 [0.7408 ; 1.1852] Additivity
    N1 Ray
    13 0.91 0.9704 [0.6774 ; 1.3900] Additivity
    N3 Ray
    15 0.93 0.8492 [0.624 ; 1.1551] Additivity
    N2 Ray
    15 0.94 1.2578 [0.9872 ; 1.6024] Additivity
  • FIG. 17 shows an isobologram of growth inhibition of UM-UC-3 cells treated with the indicated amounts of Compound (1) in combination with Compound (4) (FIG. 17A) or Compound (5) (FIG. 17B), compared to additive growth inhibition. As shown in Tables 50 and 52, and FIG. 17, Compound (1) in combination with Compound (4) or Compound (5) showed additivity on UM-UC-3 cells.
  • While there have been shown and described what are at present considered the preferred embodiments of the invention, those skilled in the art may make various changes and modifications which remain within the scope of the appended claims.

Claims (22)

  1. A method of treating a subject, wherein the subject has cancer, comprising administering to the subject an effective amount of Compound (1)
    Figure imgb0021
    or a pharmaceutically acceptable salt thereof and Compound (4)
    Figure imgb0022
    or a pharmaceutically acceptable salt thereof.
  2. The method of claim l, wherein Compound (1) and/or Compound (4) are administered with a pharmaceutically acceptable carrier.
  3. The method of claim 1, wherein Compound (1) and Compound (4) are administered in separate dosage forms.
  4. The method of claim 1, wherein Compound (1) and Compound (4) are administered in a single dosage form.
  5. The method of claim 1, wherein Compound (1) and Compound (4) are administered at different times.
  6. The method of claim 1, wherein Compound (1) and Compound (4) are administered at substantially the same time.
  7. The method of any one or claims 1-6, wherein the cancer is a mutant Kras expressing colorectal cancer.
  8. The method of any of claims 1-6, wherein said cancer is selected from the group consisting of: mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  9. The method of claim 8, wherein the effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  10. A combination for use in treating cancer, the combination comprising a therapeutically effective amount of Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof.
  11. The combination of claim 10, wherein Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof, are in separate dosage forms.
  12. The combination of claim 11, wherein Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof, are in a single dosage form.
  13. The method of any one or claims 10-12, wherein the cancer is a mutant Kras expressing colorectal cancer.
  14. The method of any one of claims 10-12, wherein said cancer is selected from the group consisting of: mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  15. The method of claim 14, wherein the therapeutically effective amount achieves a synergistic effect in the treatment of the subject in need thereof.
  16. A kit comprising: (A) Compound (1), or a pharmaceutically acceptable salt thereof; (B) Compound (4), or a pharmaceutically acceptable salt thereof; and (C) instructions for use.
  17. Use of a combination comprising a therapeutically effective amount of Compound (1), or a pharmaceutically acceptable salt thereof, and Compound (4), or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in treatment of a cancer selected from the group consisting of: mutant Kras expressing colorectal cancer, mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  18. A composition comprising a compound of the formula (1):
    Figure imgb0023
    or a pharmaceutically acceptable salt thereof, and a compound of the formula (4):
    Figure imgb0024
    or a pharmaceutically acceptable salt thereof.
  19. The composition of claim 18, further comprising a pharmaceutically acceptable carrier.
  20. The composition of claim 18, wherein said compound of formula (1) and said compound of formula (4) are present in amounts that produce a synergistic effect in the treatment of cancer in a subject in need thereof.
  21. The composition of claim 20, wherein said cancer is selected from the group consisting of mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
  22. A method of treating cancer in a subject in need thereof, comprising administering to the subject an effective amount of Compound (1)
    Figure imgb0025
    or a pharmaceutically acceptable salt thereof, and Compound (4)
    Figure imgb0026
    or a pharmaceutically acceptable salt thereof, wherein the cancer is selected from the group consisting of: mutant Kras expressing colorectal cancer, mutant Nras expressing melanoma, mutant Nras expressing bladder cancer, and mutant Braf expressing melanoma.
EP13305008.8A 2013-01-07 2013-01-07 Compositions and methods using hdm2 antagonist and mek inhibitor Withdrawn EP2752191A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP13305008.8A EP2752191A1 (en) 2013-01-07 2013-01-07 Compositions and methods using hdm2 antagonist and mek inhibitor
PCT/US2014/010461 WO2014107713A1 (en) 2013-01-07 2014-01-07 Combination of a hdm2 antagonist and a mek inhibitor for use in treating cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP13305008.8A EP2752191A1 (en) 2013-01-07 2013-01-07 Compositions and methods using hdm2 antagonist and mek inhibitor

Publications (1)

Publication Number Publication Date
EP2752191A1 true EP2752191A1 (en) 2014-07-09

Family

ID=47559359

Family Applications (1)

Application Number Title Priority Date Filing Date
EP13305008.8A Withdrawn EP2752191A1 (en) 2013-01-07 2013-01-07 Compositions and methods using hdm2 antagonist and mek inhibitor

Country Status (2)

Country Link
EP (1) EP2752191A1 (en)
WO (1) WO2014107713A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017037579A1 (en) * 2015-08-28 2017-03-09 Novartis Ag Mdm2 inhibitors and combinations thereof
US10654853B2 (en) 2016-03-04 2020-05-19 Mission Therapeutics Limited Spiro-condensed pyrrolidine derivatives as deubiquitylating enzymes (DUB) inhibitors

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201311888D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compounds
GB201311891D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compound
MX2017009454A (en) 2015-01-20 2017-10-20 Arvinas Inc Compounds and methods for the targeted degradation of the androgen receptor.
US20170327469A1 (en) 2015-01-20 2017-11-16 Arvinas, Inc. Compounds and methods for the targeted degradation of androgen receptor
TWI711452B (en) 2015-02-20 2020-12-01 日商第一三共股份有限公司 Combination therapy for cancer
US20180147202A1 (en) 2015-06-05 2018-05-31 Arvinas, Inc. TANK-BINDING KINASE-1 PROTACs AND ASSOCIATED METHODS OF USE
US10772962B2 (en) 2015-08-19 2020-09-15 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of bromodomain-containing proteins
KR102570992B1 (en) 2016-11-01 2023-08-28 아비나스 오퍼레이션스, 인코포레이티드 Tau-Protein Targeting PROTAC and Related Methods of Use
IL266842B (en) 2016-12-01 2022-09-01 Arvinas Operations Inc Tetrahydronaphthalene and tetrahydroisoquinoline derivatives as estrogen receptor degraders
US10723717B2 (en) 2016-12-23 2020-07-28 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of rapidly accelerated fibrosarcoma polypeptides
WO2018118598A1 (en) 2016-12-23 2018-06-28 Arvinas, Inc. Compounds and methods for the targeted degradation of fetal liver kinase polypeptides
BR112019012682A2 (en) 2016-12-23 2019-12-17 Arvinas Operations Inc chimeric molecules targeting egfr proteolysis and associated methods of use
US11173211B2 (en) 2016-12-23 2021-11-16 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of rapidly accelerated Fibrosarcoma polypeptides
US11191741B2 (en) 2016-12-24 2021-12-07 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of enhancer of zeste homolog 2 polypeptide
JP7266526B6 (en) 2017-01-26 2024-02-15 アルビナス・オペレーションズ・インコーポレイテッド Estrogen receptor proteolytic modulators and related methods
WO2019099926A1 (en) 2017-11-17 2019-05-23 Arvinas, Inc. Compounds and methods for the targeted degradation of interleukin-1 receptor-associated kinase 4 polypeptides
WO2019195609A2 (en) 2018-04-04 2019-10-10 Arvinas Operations, Inc. Modulators of proteolysis and associated methods of use
EP3841100A1 (en) 2018-08-20 2021-06-30 Arvinas Operations, Inc. Proteolysis targeting chimeric (protac) compound with e3 ubiquitin ligase binding activity and targeting alpha-synuclein protein for treating neurodegenerative diseases
WO2023196412A1 (en) * 2022-04-06 2023-10-12 Nobias Therapeutics, Inc. Liquid formulations comprising mitogen-activated protein kinase kinase (mek) inhibitors and methods using same

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2005121142A1 (en) 2004-06-11 2005-12-22 Japan Tobacco Inc. 5-amino-2,4,7-trioxo-3,4,7,8-tetrahydro-2h-pyrido’2,3-d! pyrimidine derivatives and related compounds for the treatment of cancer
WO2006045514A1 (en) 2004-10-20 2006-05-04 Applied Research Systems Ars Holding N.V. 3-arylamino pyridine derivatives
WO2008005266A2 (en) * 2006-06-30 2008-01-10 Schering Corporation Method of using substituted piperidines that increase p53 activity
US20100004233A1 (en) 2006-02-09 2010-01-07 Iikura Hitoshi Novel coumarin derivative having antitumor activity
WO2012065022A2 (en) 2010-11-12 2012-05-18 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2005121142A1 (en) 2004-06-11 2005-12-22 Japan Tobacco Inc. 5-amino-2,4,7-trioxo-3,4,7,8-tetrahydro-2h-pyrido’2,3-d! pyrimidine derivatives and related compounds for the treatment of cancer
WO2006045514A1 (en) 2004-10-20 2006-05-04 Applied Research Systems Ars Holding N.V. 3-arylamino pyridine derivatives
US20100004233A1 (en) 2006-02-09 2010-01-07 Iikura Hitoshi Novel coumarin derivative having antitumor activity
WO2008005266A2 (en) * 2006-06-30 2008-01-10 Schering Corporation Method of using substituted piperidines that increase p53 activity
WO2012065022A2 (en) 2010-11-12 2012-05-18 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY
ARONOV ET AL., J.MED. CHEM., vol. 52, 2009, pages 6362 - 6368
BALADULA ET AL., INVEST NEW DRUGS, vol. 29, 2011, pages 1114 - 1121
CHOU, T. C.; TALALAY, P., ADV. ENZYME REGUL., vol. 22, 1984, pages 27 - 55
D. B. SOLIT ET AL., NATURE, vol. 439, 2006, pages 358 - 362
HAURA ET AL., CLIN CANCER RES, vol. 16, no. 8, 2010, pages 2450 - 2457
HOLFORD, N. H. G.; SCHEINER, L. B., CLIN. PHARMACOKINET., vol. 6, 1981, pages 429 - 453
J. YOON ET AL: "MEK1/2 Inhibitors AS703026 and AZD6244 May Be Potential Therapies for KRAS Mutated Colorectal Cancer That Is Resistant to EGFR Monoclonal Antibody Therapy", CANCER RESEARCH, vol. 71, no. 2, 15 January 2011 (2011-01-15), pages 445 - 453, XP055058022, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-10-3058 *
JI ZHENYU ET AL: "p53 rescue through HDM2 antagonism suppresses melanoma growth and potentiates MEK inhibition.", THE JOURNAL OF INVESTIGATIVE DERMATOLOGY FEB 2012, vol. 132, no. 2, February 2012 (2012-02-01), pages 356 - 364, XP002694638, ISSN: 1523-1747 *
LOEWE, S.; MUISCHNEK, H., ARCH. EXP. PATHOL PHARMACOL., vol. 114, 1926, pages 313 - 326
R. STRAETEMANS, BIOMETRICAL JOURNAL, vol. 47, 2005
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
SOLIT DAVID B ET AL: "BRAF mutation predicts sensitivity to MEK inhibition", NATURE: INTERNATIONAL WEEKLY JOURNAL OF SCIENCE, NATURE PUBLISHING GROUP, UNITED KINGDOM, vol. 439, no. 7074, 19 January 2006 (2006-01-19), pages 358 - 362, XP002547208, ISSN: 0028-0836, DOI: 10.1038/NATURE04304 *
ZHANG ET AL., BIOOR. MED. CHEM. LET., vol. 10, 2000, pages 2825 - 2828
ZHANG WEIGUO ET AL: "MEK inhibitor AZD6244 induces cell growth arrest and synergizes nutlin-3a-mediated cell death by upregulating p53 and PUMA levels in acute myelogenous leukemia", BLOOD, vol. 110, no. 11, Part 1, November 2007 (2007-11-01), & 49TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; ATLANTA, GA, USA; DECEMBER 08 -11, 2007, pages 201A, XP002694639, ISSN: 0006-4971 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017037579A1 (en) * 2015-08-28 2017-03-09 Novartis Ag Mdm2 inhibitors and combinations thereof
IL257015A (en) * 2015-08-28 2018-03-29 Novartis Ag Mdm2 inhibitors and combinations thereof
AU2016314082B2 (en) * 2015-08-28 2019-07-25 Novartis Ag Mdm2 inhibitors and combinations thereof
AU2019204938B2 (en) * 2015-08-28 2020-07-16 Novartis Ag Mdm2 inhibitors and combinations thereof
IL277239A (en) * 2015-08-28 2020-10-29 Novartis Ag Mdm2 inhibitors and combinations thereof
EP4241850A3 (en) * 2015-08-28 2023-11-08 Novartis AG Mdm2 inhibitors and combinations thereof
US10654853B2 (en) 2016-03-04 2020-05-19 Mission Therapeutics Limited Spiro-condensed pyrrolidine derivatives as deubiquitylating enzymes (DUB) inhibitors

Also Published As

Publication number Publication date
WO2014107713A1 (en) 2014-07-10

Similar Documents

Publication Publication Date Title
EP2752191A1 (en) Compositions and methods using hdm2 antagonist and mek inhibitor
Boss et al. Clinical evaluation of AZD1152, an iv inhibitor of Aurora B kinase, in patients with solid malignant tumors
US20200368235A1 (en) Ibrutinib combination therapy
Cronin et al. Safety of triazole antifungal drugs in patients with cancer
Jhaveri et al. Heat shock protein 90 inhibitors in the treatment of cancer: current status and future directions
Reyes et al. Sorafenib and 2-deoxyglucose synergistically inhibit proliferation of both sorafenib-sensitive and-resistant HCC cells by inhibiting ATP production
Lin et al. Metformin sensitizes anticancer effect of dasatinib in head and neck squamous cell carcinoma cells through AMPK-dependent ER stress
Chern et al. Adaptive response of resistant cancer cells to chemotherapy
Wong et al. Lonafarnib for cancer and progeria
EP3137117A1 (en) Combination therapies targeting mitochondria for cancer therapy
CN105338980A (en) Pharmaceutical combinations
Gupta et al. CDK inhibitors as sensitizing agents for cancer chemotherapy
CN105050623A (en) Combination of PI3K inhibitor and c-Met inhibitor
EP3053578B1 (en) Combination cancer therapy using azabicyclo compound
EP2648729A1 (en) Compositions comprising a pi3k inhibitor and a mek inhibitor and their use for treating cancer
WO2021116929A1 (en) Composition comprising the bh3-mimetic abt-199 (venetoclax) and at least one inhibitor of the nadph production
Jabbour et al. Nilotinib for the treatment of chronic myeloid leukemia: An evidence-based review
Bose et al. Phase I trial of the combination of flavopiridol and imatinib mesylate in patients with Bcr-Abl+ hematological malignancies
Schmukler et al. Chloroquine synergizes with FTS to enhance cell growth inhibition and cell death
AU2012307309A1 (en) Compositions and methods for treating cancer using PI3K beta inhibitor and MAPK pathway inhibitor, including MEK and RAF inhibitors
MX2008014292A (en) Combination comprising an iron chelator and an anti-neoplastic agent and use thereof.
KR102607967B1 (en) Combination of Protein Kinase Inhibitors and Additional Chemotherapeutic Agents
US20190105323A1 (en) Tyrosine kinase inhibitors for use in a method of treating cancer in association with a reduced caloric intake
Cornely et al. Numbers needed to treat with posaconazole prophylaxis to prevent invasive fungal infection and death
Failly et al. Combination of sublethal concentrations of epidermal growth factor receptor inhibitor and microtubule stabilizer induces apoptosis of glioblastoma cells

Legal Events

Date Code Title Description
17P Request for examination filed

Effective date: 20130107

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

R17P Request for examination filed (corrected)

Effective date: 20150109

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20150110